DI(2-ETHYLHEXYL)PHTHALATE
                                                 o
                                                 o
Agency for Toxic Substances and Disease Registry
U.S. Public Health Service

-------
                                                       ATSDR/TP-88/15
           TOXICOLOGICAL PROFILE FOR
           DI(2-ETHYLHEXYL)PHTHALATE
             Date Published — April 1989
                    Prepared by:

                  Life Systems, Inc.
            under Contract No. 68-02-4228

                        for

Agency for Toxic Substances and Disease Registry (ATSDR)
              U.S. Public Health Service

                 in collaboration with

      U.S. Environmental Protection Agency (EPA)
       Technical editing/document preparation by:

            Oak Ridge National Laboratory
                       under
     DOE Interagency Agreement No. 1857-B026-A1

-------
                          DISCLAIMER

Mention of company name or product does not constitute endorsement by
the Agency for Toxic Substances and Disease Registry.

-------
                                FOREWORD

      The  Superfund Amendments and Reauthorization Act  of  1986  (Public
 Law 99-499) extended and amended the Comprehensive  Environmental
 Response, Compensation, and Liability Act of 1980 (CERCLA or Superfund)
 This public law  (also known as SARA) directed the Agency  for Toxic
 Substances and Disease Registry (ATSDR) to prepare  toxicological
 profiles  for hazardous substances which are most commonly found at
 facilities on the CERCLA National Priorities List and  which pose  the
 most significant potential threat to human health,  as  determined  by
 ATSDR and the Environmental Protection Agency (EPA). The  list  of  the 100
 most significant hazardous substances was published in the Federal
 Register  on April 17, 1987.

      Section 110 (3) of SARA directs the Administrator of ATSDR to
 prepare a toxicological profile for each substance  on  the list. Each
 profile must include the following content:

      "(A)  An examination,  summary, and interpretation of available
      toxicological information and epidemiologic evaluations on a
      hazardous substance in order to ascertain the  levels of significant
      human exposure for the substance and the associated  acute,
      subacute, and chronic health effects.

      (B)  A determination of whether adequate information on the  healch
      effects of each substance is available or in the  process  of
      development to determine levels of exposure which present a
      significant risk to human health of acute,  subacute,  and  chronic
      health effects.

      (C)  Where appropriate,  an identification of toxicological testing
      needed to identify the types or levels of exposure that may  present
      significant risk of adverse health effects in  humans."

      This toxicological profile is prepared in accordance with
 guidelines developed by ATSDR and EPA.  The guidelines  were published in
 the Federal Register on April 17,  1987.  Each profile will be revised and
 republished as necessary,  but no less often than every three years, as
 required by SARA.

     The ATSDR toxicological  profile is  intended to characterize
 succinctly the toxicological  and health  effects  information for the
hazardous substance  being  described  Each profile identifies and  reviews
 the key literature  that describes  a hazardous substance's  toxicological
properties.  Other literature  is presented but described in less detail
 than the key studies.  The  profile  is not intended to be an exhaustive
document;  however, more comprehensive sources of specialty information
are referenced.
                                                                      111

-------
 foreword


      Each toxicological profile begins  with a public health statement,
 which describes In nontechnical language  a substance's relevant
 toxicological properties.  Following the statement Is material that
 presents levels of significant human exposure and,  where known,
 significant health effects.  The adequacy  of Information to determine  a
 substance's health effects Is  described in a health effects summary.
 Research gaps in toxicologic and health effects  information are
 described in the profile.  Research  gaps that are of significance to
 protection of public health  will be Identified by ATSDR,  the National
 Toxicology Program of the  Public Health Service,  and EPA.  The focus of
 the profiles is on health  and  toxicological information;  therefore, we
 have included this information in the front of the  document.
      The principal audiences for the  toxicological  profiles are health
 professionals at the federal,  state,  and  local levels,  interested
 private  sector organizations and groups,  and members of the public. We
 plan to  revise these documents in response  to public comments and as
 additional data become  available; therefore,  we  encourage  comment that
 will make the toxicological  profile series  of the greatest use.

      This profile  reflects our assessment of all  relevant  toxicological
 testing  and information that has been peer  reviewed.  It has been
 reviewed by scientists  from  ATSDR,  EPA, the  Centers  for Disease Control,
 and the  National Toxicology  Program. It has  also  been reviewed by a
 panel of nongovernment  peer  reviewers and was  made  available  for public
 review.  Final responsibility for the contents  and views expressed in
 this toxicological profile resides with ATSDR.
                                    James 0. Mason, M.D.,  Dr.  P.H.
                                    Assistant Surgeon  General
                                    Administrator, ATSDR
iv

-------
                                CONTENTS

FOREWORD 	  llt

LIST OF FIGURES 	   ix

LIST OF TABLES 	   xi

 1.   PUBLIC HEALTH STATEMENT  	    1
     1.1  WHAT IS  DI(2-ETHYLHEXYL)PHTHALATE?  	'.'.'.'.'.'.'.'.'.'    1
     1.2  HOW MIGHT I  BE  EXPOSED TO DI(2-ETHYLHEXYL)PHTHALATE?  	    1
     1.3  HOW DOES DI(2-ETHYLHEXYL)PHTHALATE  GET  INTO MY BODY?  ...     2
     1.4  HOW CAN  DI (2 - ETHYLHEXYL) PHTHALATE AFFECT MY HEALTH?  	    2
     1.5  IS THERE A MEDICAL  TEST TO DETERMINE  IF I HAVE
          BEEN EXPOSED TO DI(2-ETHYLHEXYL)PHTHALATE?  	    2
     1.6  WHAT LEVELS  OF  EXPOSURE HAVE RESULTED IN HARMFUL
          HEALTH EFFECTS? 	    3
     1.7  WHAT RECOMMENDATIONS HAS THE FEDERAL  GOVERNMENT MADE	
          TO PROTECT HUMAN HEALTH? 	    3
 2.   HEALTH EFFECTS SUMMARY 	        7
     2.1  INTRODUCTION 	'.'.'.'.'.'.'.'.'.'..    7
     2.2  LEVELS OF SIGNIFICANT  HUMAN EXPOSURE  	'.'.'.'.'.'.'.'.'.'.    8
          2.2.1  Key Studies  and Graphic Presentations  	     8
                2.2.1.1  Lethality/decreased longevity  	    8
                2.2.1.2  Systemic/target organ toxicity 	   13
                2.2.1.3  Developmental toxicity  	   15
                2.2.1.4  Reproductive toxicity	 . . .   16
                2.2.1.5  Genotoxicity 	   17
                2.2.1.6  Carcinogenicity 	   18
          2.2.2  Biological Monitoring as a Measure of Exposure
                and Effec ts  	   21
          2.2.3  Environmental Levels as Indicators of Exposure
                and Effects  	   22
                2.2.3.1  Levels found in the environment 	    22
                2.2.3.2  Human  exposure potential 	    22
     2.3  ADEQUACY OF DATABASE 	   22
         2.3.1  Introduction 	   22
         2.3.2  Health Effect End Points 	'.'.'.'.'.   23
                2.3.2.1  Introduction and graphic summary 	   23
                2.3.2.2  Description of highlights of graphs ....    23
                2.3.2.3  Summary of  relevant ongoing research  ....   23
         2.3.3  Other Information Needed for Human
                Health Assessment  	    26
                2.3.3.1  Pharmacokinetics and mechanisms
                         of action . .     	   26
                2.3.3.2  Monitoring  of human biological samples  ..   26
                2.3.3.3  Environmental considerations 	     26

-------
 Concents

  3.  CHEMICAL AND PHYSICAL INFORMATION 	                  29
      3.1  CHEMICAL IDENTITY	' ' '	   29
      3 . 2  CHEMICAL AND PHYSICAL PROPERTIES 	'.'.'.'.'.'.   29
  4.  TOXICOLOGICAL DATA 	        33
      4.1  OVERVIEW 	   	   33
      4.2  TOXICOKINETICS 	'.'.'.'.'.'.'.'.'.'.'.'.'.'.'.'.   34
           4.2.1  Absorption 	   34
                  4.2.1.1  Inhalation 	'.'.'.'.'.'.'.'.'.        34
                  4.2.1.2  Oral 	   34
                  4.2.1.3  Dermal 	'.'.'.'.'.   35
           4.2.2  Distribution 	   35
                  4.2.2.1  Inhalation 	 "       36
                  4.2.2.2  Oral 	   36
                  4.2.2.3  Dermal 	   38
                  4.2.2.4  Systemic  	   38
           4.2.3  Metabolism	   39
                  4.2.3.1  Inhalation 	'.'.'.'.'.'.'.'.	   39
                  4.2.3.2  Oral 	"   4!
                  4.2.3.3  Dermal 	'.'.'.'.'.   42
                  4.2.3.4  In vitro  studies 	   43
           4.2.4  Excretion 	   43
                  4.2.4.1  Inhalation 	'.'.'.	   43
                  4.2.4.2  Oral 	   43
                  4.2.4.3  Dermal 	'//_   45
                  4.2.4.4  Systemic  	   45
           4.2.5  Discussion 	           45
      4.3   TOXICITY 	'.'.'.'.'.'.'.'.'.'.'.'.'.'.'.'.   47
           4.3.1  Lethality and Decreased  Longevity  	   47
                  4.3.1.1  Inhalation 	           47
                  4.3.1.2  Oral 	   47
                  4.3.1.3  Dermal  	   49
                  4.3.1.4  Discussion 	   49
           4.3.2  Systemic/Target Organ Toxicity  	   49
                  4.3.2.1  Hepatic toxicity 	   49
                  4.3.2.2  Testicular toxicity  	   53
           4.3.3  Developmental Toxicity 	   53
                  4.3.3.1  Inhalation 	     53
                  4.3.3.2   Oral 	\\\   53
                  4.3.3.3   Dermal  	   55
                  4.3.3.4  Discussion 	   55
           4.3.4  Reproductive  Toxicity 	   56
                  4.3.4.1   Inhalation 	   56
                  4.3.4.2  Oral 	'.'.'.'.'.   56
                  4.3.4.3  Dermal  	   60
                  4.3.4.4  General discussion 	   60
           4.3.5   Genotoxicity  	   61
                  4.3.5.1  Genotoxicity studies 	   61
                  4.3.5.2  Discussion 	   61
           4.3.6   Carcinogenicity  	   64
                  4.3.6.1  Inhalation 	   64
                  4.3.6.2  Oral  	   64
                  4.3.6.3  Dermal 	   67
vi

-------
                                                                Concents

                 4. 3.6.4  Possible mechanisms of carcinogenesis ...   68
                 4.3.6.5  General  discussion 	         76
     4.4  INTERACTION WITH OTHER CHEMICALS 	\   75
 5.   MANUFACTURE,  IMPORT,  USE,  AND DISPOSAL 	      77
     5.1  OVERVIEW 	    '     77
     5.2  PRODUCTION 	                       77
     5.3  IMPORT 	                    	   77
     5.4  USE 	'.'.'.'..'.'.'."   78
     5.5  DISPOSAL 	   78

 6.   ENVIRONMENTAL FATE  	              79
     6.1  OVERVIEW 	'.'.'.'.'.'.'.'.'.'.'.   79
     6 . 2  RELEASES TO  THE  ENVIRONMENT  	'.'.'.'.'.'.'.'.'.'.'.   79
          6.2.1  Anthropogenic  Sources  	   79
          6.2.2  Natural Sources 	                     79
     6 . 3  ENVIRONMENTAL  FATE  	'.'.'.'.'.'.'.   81
          6.3.1  Atmospheric  Fate  Processes  	   81
          6.3.2  Surface Water/Groundwater Fate  Processes  	   81
          6.3.3  Soil  Fate  Processes 	   82
          6.3.4  Biotic  Fate  Processes  	   82
 7.   POTENTIAL FOR HUMAN EXPOSURE  	            83
     7.1  OVERVIEW 	'   83
     7.2  LEVELS MONITORED  OR ESTIMATED IN THE ENVIRONMENT  ........   83
          7.2.1  Levels  in  Air  	   83
          7.2.2  Levels  in  Water 	   84
          7.2.3  Levels  in  Soil  	   85
          7.2.4  Levels  in  Food  	   85
          7.2.5  Resulting  Exposure Levels  	   85
     7.3   OCCUPATIONAL EXPOSURES 	     86
     7.4   POPULATIONS AT HIGH RISK  	   86
          7.4.1  Above-Average Exposure 	   86
          7.4.2  Above-Average Sensitivity  	   87
8.   ANALYTICAL METHODS  	   89
    8.1   Environmental Media 	   89
          8.1.1  Air 	'.'.'.'.'.'.'.'.'.   89
          8.1.2  Water 	       89
          8.1.3  Soil  	.......   91
          8.1.4  Food  	'   91
    8 .2  BIOMEDICAL SAMPLES 	'.'.'.'.'.'.'.'.'.   91
         8.2.1  Fluids and Exudates 	   91
         8.2.2  Tissues 	   91

9.   REGULATORY AND ADVISORY STATUS 	        93
    9 .1  INTERNATIONAL  	' '   93
    9 . 2  NATIONAL 	' ' ."    93
         9.2.1  Regulations 	     93
         9.2.2  Advisory Guidance  	     95
         9.2.3  Data Analysis 	       95
                9.2.3.1  Reference dose 	      95
                9.2.3.2  Carcinogenic  potency 	     96
                                                                    VI 1

-------
 Concents

       9.3   STATE  	      97
            9.3.1  Regulations	   97
            9.3.2  Advisory Guidance  	 //   97

 10.   REFERENCES  	         9g

 11.   GLOSSARY  	         115

       APPENDIX:   PEER REVIEW	 119
viii

-------
                            LIST OF FIGURES
1.1  Health effects from breathing di(2-ethylhexyDphthalate 	    4
1.2  Health effects from Ingesting dl(2-ethylhexyl)phthalate 	    5
2.1  Effects of dl(2-ethylhexyl)phthalate--inhalatlon exposure 	    9
2.2  Effects of di(2-ethylhexyDphthalate--oral exposure 	   10
2.3  Levels of significant exposure for di(2-ethylhexyl)-
     phthalate--Inhalation 	   H
2.4  Levels of significant exposure for di(2-ethylhexyl)-
     phthalate--oral 	   12
2.5  Availability of information on health effects of
     dl(2-ethylhexyl)phthalate (human data) 	   24
2.6  Availability of information on health effects of
     di(2-ethylhexyl)phthalate (animal data) 	   25
4.1  Proposed metabolism for di(2-ethylhexyl)phthalate
     in the rat 	   40
4.2  Species-specific metabolism of di(2-ethylhexyl)phthalate 	   46

-------
                             LIST OF TABLES
2.1  Summary of ongoing research 	    27
3.1  Chemical identity of di(2-ethylhexyl)phthalate 	   30
3.2  Physical and chemical properties of
     di(2-ethylhexyl)phthalate 	   31
4.1  Acute lethality of di(2-ethylhexyl)phthalate 	   48
4.2  Results of short-term assays 	   62
4.3  Incidence of hepatocellular carcinomas and neoplastic
     nodules observed in DEHP-dosed rats 	   66
4.4  Incidence of hepatocellular carcinomas and hepatocellular
     adenomas observed in DEHP-dosed mice 	   66
6.1  Releases of di(2-ethylhexyl)phthalate to the environment 	   80
8.1  Analytical methods for di(2-ethylhexyl)phthalate
     in environmental samples 	    90
8.2  Analytical methods for di(2-ethylhexyl)phthalate
     in biological samples 	    92
9.1  Regulations and guidelines applicable to
     di(2-ethylhexyl)phthalate 	     94
                                                                      XI

-------
                      1.  PUBLIC HEALTH STATEMENT

1.1  WHAT IS DI(2-ETHYLHEXYL)FHTHALATE?

     Di(2-ethylhexyl)phthalate,  commonly referred Co as DEHP, is a
liquid widely used to make plastics more flexible. Plastics may contain
from 1 to 40% DEHP by weight and are widely used in consumer products
such as imitation leather, rainwear, footwear, upholstery, flooring,
tablecloths, shower curtains, food packaging materials, and children's
toys. Plastics containing OEHP are also used for Cubing and containers
for blood transfusions and blood products.  OEHP is also used as a
hydraulic fluid and as a dielectric fluid (a nonconductor of electric
current) for use in electrical capacitors.

1.2  HOW MIGHT I BE EXPOSED TO DI(2-ETHYLHEXYL)PHTHALATE?

     About 300 million pounds of DEHP are used each year to manufacture
plastic products for commercial, medical, and consumer use. Low-level
exposures to DEHP from the air or drinking water occur whenever these
products are used.  Plasticizers  such as DEHP do not become a permanent
part of the plastic matrix during the manufacturing process. Thus, under
certain use or disposal conditions, DEHP can migrate from plastic
products into the environment. As a result, DEHP is widely distributed
in the environment, and exposure can occur via air, water, and food.
Most exposures to DEHP are at very low levels, and exposures via air and
water are expected to be minimal.

     A potential exists for DEHP contamination of food during
processing, handling, transportation, and packaging. DEHP is found in
animal products used for human consumption. The highest levels have been
detected in milk and cheese. The U.S. Food and Drug Administration (FDA)
currently allows the use of DEHP plasticized containers or wrappings
only for foods that primarily contain water; therefore, properly
packaged foods are unlikely to become contaminated. The FDA ruling,
however, does not preclude the possible misuse of DEHP plasticized
containers or wrappings by the consumer.

     Acute exposures to relatively high levels (compared to DEHP levels
commonly found in food or drinking water) of DEHP can occur when DEHP
migrates from the plastics used  in medical apparatus (such as storage
bags and tubing) used for blood  transfusions or kidney dialysis.
Patients exposed to DEHP through medical procedures such as dialysis may
conceivably be at risk from its  toxic effects, but confirmatory studies
are necessary to show this relationship.

     Some exposure may occur from the evaporation of DEHP from
plasticized products during their use.  The level of such exposures will
depend on the thickness of the plastic item, the temperature, and the
specific nature of the product.  Therefore,  it is difficult to estimate

-------
2   Section 1

the level of this kind of exposure.  However, because  the  vapor pressure
of DEHP is very low,  and essentially no  evaporation occurs  at normal
room temperature, inhalation exposure is expected  to  be minimal.
     Most discarded plasticized products are disposed of  either by
incineration or by dumping in a landfill.  Incineration is not likely  to
result in significant exposures to DEHP.  After  disposal of  products in a
landfill, the DEHP will slowly leach from the plastic product and may
reach the groundwater. However, the low  solubility of DEHP  in water
limits the amounts that will enter the environment by this  route.
     Exposure from water nay also occur  if DEHP migrates  from some kinds
of flexible plastic tubing used to carry drinking  water.  Again, because
DEHP is not very soluble in water, the exposure level may be low but
could continue for a long period of time.
     Estimation of human exposure to DEHP Is very  complex because of  the
wide range of items containing DEHP and  the large  number  of variables
influencing how much of the DEHP content of an  item would reach an
exposed individual and be absorbed into  the body.

1.3  HOW DOES DI(2-ETHYLHEXYL)PHTHALATE  GET INTO MY BODY?
     There are several potential sources' of exposure  to DEHP. Humans  are
primarily exposed through foods that come into  contact with packaging
materials containing DEHP. Once DEHP gets into  the gastrointestinal
tract, it is quickly absorbed and gets into the blood and is rapidly
metabolized. Humans may also receive higher exposures during blood
transfusions and kidney dialysis because of the movement  of DEHP into
blood from plastic bags and tubing.

1.4  HOW CAN DI(2-ETHYLHEXYL)FHTHALATE AFFECT MY HEALTH?
     There are essentially no studies on the health effects of DEHP in
humans. However, DEHP causes cancer in rats and mice. It  is also known
to produce liver damage and male reproductive  system damage, affect
reproduction, and produce birth defects  in laboratory animals. Because
none of these effects have been documented in humans, it  is difficult to
estimate the kinds of health effects and exposure levels  that may
actually affect humans. However, it is prudent  to regard the animal data
as indicating some degree of concern for harmful human effects until
research can more reasonably conclude that no  harm can occur.

1.5  IS THERE A MEDICAL TEST TO DETERMINE IF  I  HAVE
     BEEN EXPOSED TO DI(2-ETHYLH£XYL)PHTHALATE?
     The major method for determining whether  a person has been  exposed
to DEHP is by testing the urine. After exposure to DEHP,  the body
metabolizes it into two major chemical products. mono(2-ethylhexyl)-
phthaiate (MEHP) and 2-ethylhexanol. Detection of DEHP and/or  these
chemicals in the urine will  indicate whether  a person has been recently
exposed. In addition, the blood can be analyzed for DEHP, MEHP,  and  2-
ethylhexanol shortly after exposure.

-------
                                            Public  Health  Statement   3

L.6  WHAT LEVELS OF EXPOSURE HAVE  RESULTED  IN
     HARMFUL HEALTH EFFECTS?
     The graphs on Che following pages  (Figs.  1.1  and 1.2)  show the
relationship between exposure to DEHP and known health effects.  In the
first set of graphs (Fig.  1.1).  labeled "Health effects from breathing
DEHP - exposure is measured in milligrams of DEHP  per cubic meter of  air
(me/m3). In all graphs,  effects In animals  are shown on the left side,
and effects in humans are shown on the  right.  In the second set of
graphs (Fig  1.2), the same relationship is represented for the known
"Health effects from ingesting DEHP." Exposures are  measured in
milligrams of DEHP per kilogram of body weight (mg/kg).
     Figure 1 1 summarizes the effects  resulting from short-term
inhalation of DEHP at levels up co -24,000  mg/m3.  No adverse effects
were observed in the studies that were reviewed, and no longer-term
inhalation studies have been done. Figure 1.2  shows  the general types of
adverse health effects resulting from ingestion of DEHP at various
exposure levels. In short-term animal studies, death has resulted from
ingestion of relatively high doses of DEHP. In other animal studies,
both short-term and long-term ingestion of DEHP has resulted in liver
toxicity, reproduction toxicity, and toxicity to the unborn or newborn
These effects have been observed only in animal studies. Similar
information based on human  exposures to DEHP  is not available.
     DEHP has also been shown to cause liver  tumors in both rats  and
mice receiving  DEHP in their diet  throughout  their entire  life  span.
Based on the results  of these cancer studies,  the excess risk  to  humans
has been estimated. The number  of  excess cases  of cancer which  would be
produced in populations of  10,000, 100.000, 1.000,000, or  10,000,000
individuals exposed to 1 microgram of DEHP  per  liter  for their  entire
lifetime is estimated to be 0.024. 0.24, 2.4,  and 24.  respectively.  It
should  be noted that  these  risk values  are  plausible  upper-limit
estimates. Actual risk levels  are  unlikely  to be higher and may be
lower.

1.7   WHAT RECOMMENDATIONS  HAS  THE FEDERAL  GOVERNMENT
      MADE TO  PROTECT  HUMAN HEALTH?
      The federal government has taken  a number of steps  to protect
humans  from DEHP. The Environmental  Protection Agency (EPA) has
authority to regulate DEHP under  the Resource Conservation and Recovery
Act (RCRA)-  the Comprehensive Environmental Response. Compensation,  and
 Liability Act (CERCLA or "Superfund");  the Solid Waste Disposal Act; the
 Clean Water Act; the Safe  Drinking Water Act; the Clean Air Act; and the
 Toxic Substances Control Act.  The EPA has  identified DEHP as a toxic
waste material and requires that  persons who  generate, transport  treat.
 store,  or dispose of it comply with  regulations of  the EPA Federal
 Hazardous Waste Management Program.  The EPA has also identified DEHP as
 a hazardous constituent of wastes from the cleaning equipment and tanks
 used in paint manufacturing. The  EPA has established human health
 criteria for ingestion of water and contaminated aquatic  organisms  (such
 as fish and shellfish) at  15 milligrams per  liter of water for
 noncancerous health effects. EPA has also calculated risk estimates for
 the probability of developing cancer from  ingesting DEHP. The

-------
      Section  1
        SHORT-TERM EXPOSURE
    (LESS THAN OR EQUAL TO 14 DAYS)
                                         LONG-TERM EXPOSURE
                                        (GREATER THAN 14 DAYS)
  EFFECTS
     IN
  ANIMALS
NO LETHALITY-
CONG. IN
  AIR
 (mg/m3)

 100.000
               10.000
               1.000
                100
                10
  EFFECTS
     IN
  HUMANS


QUANTITATIVE
DATA WERE
NOT AVAILABLE
EFFECTS CONC. IN EFFECTS
IN AIR IN
ANIMALS (mg/m3) HUMANS
QUANTITATIVE 100
DATA WERE
NOT AVAILABLE
10.
1.C
1C
1
,000 QUANTITATIVE
DATA WERE
NOT AVAILABLE
300
00
0
3
               Fig. 1.1. Health effects from breathing di(2-etbylhexyl)plithalate.

-------
                                                        Public Health Statement
            SHORT-TERM EXPOSURE
        (LESS THAN OR EQUAL TO 14 DAYS)
                                    LONG-TERM EXPOSURE
                                   (GREATER THAN 14 DAYS)
      EFFECTS
         IN
      ANIMALS
     DEATH•
REPRODUCTIVE
   TOXICITY
      LIVER
    TOXICITY
TOXIC EFFECTS
ON UNBORN OR
   NEWBORN
  DOSE
(mg/kg/day)

 100.000

-^<

— ^«
10.(

I — 1.C
1(

00

00
0
   EFFECTS
      IN
   HUMANS

QUANTITATIVE
DATA WERE
NOT AVAILABLE
EFFECTS
   IN
ANIMALS
  DOSE
(mg/kg/day)

 100.000
EFFECTS
   IN
HUMANS
                     10
                                                                       QUANTITATIVE
                                                                       DATA WERE
                                                                       NOT AVAILABLE
                                                            10.000
                                           1.000
                      REPRODUCTIVE
                         TOXICITY	
                      TOXIC EFFECTS
                      ON UNBORN OR
                        NEWBORN
                          LIVER
                         TOXICITY
                                                              100
                                            10
                     10
                                            1 0
               Fig. 1.2. Health effects from ingesting di(2-*thylhexyl)phthalate.

-------
    Section 1
regulates the chemical as an unintentional food additive.  Finally,  the
Occupational Safety and Health Administration (OSHA)  has set a
permissible exposure level of 5 milligrams DEHP per cubic  meter of air
for occupational exposures to DEHP.

-------
                        2.   HEALTH EFFECTS SUMMARY

 2.1  INTRODUCTION

      This section summarizes and graphs data on the health effects
 concerning exposure to DEHP.  The purpose of this section is to  present
 levels of significant exposure for DEHP based on key toxicological
 studies,  epidemiological  investigations, and environmental exposure
 data.  The information presented in this section is  critically evaluated
 and discussed in Sect.  4,  Toxicological Data,  and Sect.  7,  Potential  for
 Human Exposure.

      This Health Effects  Summary section comprises  two major parts.
 Levels of Significant Exposure (Sect.  2.2)  presents brief narratives  and
 graphics  for key studies  in a manner  that provides  public health
 officials,  physicians,  and other interested individuals  and groups with
 (1)  an overall perspective of the toxicology of DEHP and (2) a
 summarized depiction of significant exposure levels associated  with
 various adverse  health  effects.  This  section also includes  information
 on  the levels  of DEHP that have  been  monitored in human  fluids  and
 tissues and information about levels  of DEHP found  in environmental
 media  and their  association with human exposures.

     The  significance of the  exposure  levels shown  on the graphs may
 differ depending on the user's perspective.  For example,  physicians
 concerned with the  interpretation of  overt  clinical findings in exposed
 persons or  with  the  identification of  persons  with  the potential to
 develop such disease  may be interested in levels  of exposure associated
 with frank  effects  (Frank  Effect Level,  FEL).  Public health officials
 and project managers  concerned with response actions at  Superfund sites
 may want  information  on levels of exposure  associated with  more subtle
 effects in  humans or  animals  (Lowest-Observed-Adverse-Effect Level,
 LOAEL)  or exposure  levels below  which  no adverse  effects  (No-Observed-
 Adverse-Effect Level, NOAEL) have  been observed.  Estimates  of levels
 posing minimal risk to  humans  (Minimal  Risk Levels)  are  of  interest to
 health professionals  and citizens  alike.

     Adequacy of Database  (Sect.  2.3)  highlights  the availability of key
 studies on  exposure to  DEHP in the scientific  literature  and displays
 these  data  in three-dimensional  graphs  consistent with the  format in
 Sect.  2.2.  The purpose  of this section  is to suggest where  there might
be insufficient information to establish  levels of  significant human
exposure.  These areas will be considered by  the Agency for  Toxic
Substances  and Disease Registry  (ATSDR),  EPA,  and the  National
Toxicology  Program (NTP) of the U.S. Public Health  Service  in order to
develop a research agenda for DEHP.

-------
8   Section 2

2.2  LEVELS OF SIGNIFICANT HUMAN EXPOSURE

     To help public health professionals address the needs  of persons
living or working near hazardous waste sites,  the toxicology data
summarized in this section are organized first by route  of  exposure--
inhalation, ingestion, and dermal--and then by toxicological end points
that are categorized into six general areas--lethality,  systemic/target
organ toxicity, genetic toxicity, and carcinogenicity. The  data are
discussed in terms of three exposure periods--acute, intermediate, and
chronic.
     Two kinds of graphs are used to depict the data. The first.type  is
a "thermometer" graph. It provides a graphical summary of the human  and
animal toxicological end points (and levels of exposure) for each
exposure route for which data are available. The ordering of effects
does not reflect the exposure duration or species of animal tested.  The
second kind of graph shows Levels of Significant Exposure (LSE) for  each
route and exposure duration. The points on the graph showing NOAELs  and
LOAELs reflect the actual doses (levels of exposure) used in the key
studies. No adjustments for exposure duration or intermittent exposure
protocol were made.
     Adjustments reflecting the uncertainty of extrapolating animal  data
to man, intraspecies variations, and differences between experimental vs
actual human exposure conditions were considered when estimates of
levels posing minimal risk to human health were made for noncancer end
points. These minimal risk levels were derived for the most sensitive
noncancer end point for each exposure duration by applying uncertainty
factors. These levels are shown on the graphs as a broken line starting
from the actual dose (level of exposure) and ending with a concave-
curved line at its terminus. Although methods have been established to
derive these minimal risk levels  (Barnes et al. 1987), shortcomings
exist in the techniques that reduce the confidence  in the projected
estimates. Also shown on the graphs under the cancer end point are low-
level risks (10'4  to 10'7) reported by EPA. In addition, the actual dose
(level of exposure) associated with the tumor incidence is plotted.

2.2.1  Key Studies and Graphic Presentations
     Figures 2.1 and 2.2 are graphic presentations  of the adverse health
effects that have been observed  in studies  with experimental animals
using inhalation and ingestion exposure routes,  respectively.  Animal
studies using dermal exposure were not  located  in  the available
literature. Human  data on the adverse health effects of DEHP were not
available.
     Figures 2.3 and 2.4 present  data  for various  toxicity  end points
for the inhalation and oral  routes,  respectively.  Details  of these
studies are presented in  the  following  subsections.

2.2.1.1  Lethality/decreased  longevity
     No animal studies provided  appropriate data to evaluate lethality
or decreased longevity as a  toxicity end point  for DEHP exposure  via
dermal  exposure.  In  inhalation  studies  with rats,  lethality was not

-------
                                                             Health  Effects Summary     9
  ANIMALS
   (mgftn3)
                                                                     HUMANS
100.000 |—
 10.000
  1.000
   100
    10
          O RAT. ACUTE LETHALITY. 1 h
                                       O NOAEL
QUANTITATIVE DATA
WERE NOT AVAILABLE
                 Fig. 2.1  Effects of di(2-ethylhexyl)phthalate—inhalation exposure.

-------
10     Section 2
   ANIMALS
  (mg/Vg/day)
                                                                       HUMANS
100 0001-
 10.000
  100
   10
             RABBIT LDjo
             RAT. LD,o
             GUINEA PIG. LDM
             MOUSE LDM
             MOUSE. LIVER EFFECTS 14 DAYS
          O  MONKEY. TESTICULAR AND LIVER EFFECTS  14 DAYS
          •  MOUSE FEMALE. CANCER. 2 YEARS
          •  MOUSE. MALE. CANCER 2 YEARS
          •  RAT. TESTICULAR EFFECTS 14 DAYS
          •  RAT (SUCKLING). LDW 5 DAYS
          •  HAMSTER. UVER EFFECTS. 14 DAYS
          •  RAT. CANCER. 2 YEARS
      \f»  RAT. TESTICULAR EFFECTS. 2 YEARS
        \O  RAT. UVER EFFECTS. 2 YEARS
        **•  RAT. DEVELOPMENTAL EFFECTS. 20 DAYS DURING GESTATION
        ~"O  RAT. TESTICULAR EFFECTS. 2 YEARS
        ;*;•  RAT. TESTICULAH EFFECTS 60 DAYS
        ^O  HAMSTER. LIVER EFFECTS. 14 DAYS

             MOUSE. FERTILITY EFFECTS 106 DAYS (MATING AND POST MATING)
             BAT I IIJPB fff^**^t* * * M.*..dh
f • RAT. LIVER EFFECTS. 14 DAYS
\ O RAT (SUCKLING). LD^ 5 DAYS
            MOUSE DEVELOPMENTAL EFFECTS. DAYSO-17OF GESTATION
         O RAT. TESTICULAR EFFECTS. 60 DAYS
            RAT. LIVER EFFECTS. 3 AND 28 DAYS
            MOUSE. DEVELOPMENTAL EFFECTS DAY 7 OF GESTATION
            MOUSE. DEVELOPMENTAL EFFECTS. DAYSO-17OF GESTATION
        -O RAT. UVER EFFECTS. 14 DAYS
         • GUINEA PIG. LIVER EFFECTS. 1 YEAR

         O MOUSE. FERTILITY EFFECTS. 105 DAYS (MATING AND POST MATING)
                                    • LOAEL
                                    O NOAEL
                                                                            QUANTITATIVE DATA
                                                                            WERE NOT AVAILABLE
               Fig. 2.2.  Effects of dK2-ettaylhexyl)pbthmUte—oral exposure.

-------
                                                 Health Effects Summary   11
  (mg/m3)

100.000
10.000
1,000
 100
 10
                 ACUTE
               (£14 DAYS)
                LETHALITY
              r  RAT
            INTERMEDIATE
            (15-364 DAYS)
                                         QUANTITATIVE
                                         DATA WERE NOT
                                         AVAILABLE
                                                                  CHRONIC
                                                                .(2365 DAYS)
                                 QUANTITATIVE
                                 DATA WERE NOT
                                 AVAILABLE
i  MINIMAL RISK FOR EFFECTS    O NOAEL
  OTHER THAN CANCER
       Fig. 2.3. Leveb of significant exposure for di(2-ethylhexyl)phthala(e—inhalation.

-------
ACUTE INTERMEDIATE CHROMC
(iUDAYS) (15-364 DAYS) (2365 DAYS)
DEVELOP REPHO TARGET TARGET REPRO- DEVELOP- TARGET REPHO
t— •
ro

LETHALITY MENTAL DUCTON ORGAN ORGAN DUCTON MENTAL ORGAN DUCTON CANCER !?
(ing 'kg/day)
100.000 r-



10000

1.000
100
10
1
01
001

0001


•
00001
000001


1000001

9 th
|i ' Om(UVER)
~
Ok Oh (LIVER) »'
'V
O
n
K-
o
a


n F
• r (SUCKLING) «r fs (LIVER) 9mM
T T Or(UVER) •- •'
i .. « 4
6 «r(LIVER) If1" ,m
• m •MUVER) »r (LIVER) <•> T
! r
-
-
^ ,0^,
vi/
io-» -



.0--
• LOAEL g GUINEA PIG 1 HAMSTER ! MNMALRBK '^
O NOAEL m MOUSE k MONKEY j LEVEL FOR EFFECTS
h RABBIT M MALE J, OTHER THAN CANCER
r RAT F FEMALE




ESTIMATED
UI'PER BOUND
HUMAN
CANCER
niSK LfcVfLS




Fig. 2.4. Levels of significant exposure for di(2-cthylbexyl)phthalate—oral.

-------
                                             Healch Effects Summary   13

observed following a 1-h exposure at a dose level of 23,670 rng/nt^
(1,457 ppm)  (WARF Institute 1976) or a 6-h exposure at a dose level of
600 mg/m3 (37 ppm) (Pegg 1979)  No human data were available for either
of these routes of administration.
     In an acute oral toxicity study reported by Shaffer et al  (1945),
single doses of DEHP were administered by gavage to male Uistar rats.
The median lethal dose (LD50) was estimated to be 30,600 (95% confidence
limits, 20,800 to 45,200) mg/kg.
     In other acute oral toxicity studies, Shaffer et al. (1945)
reported an LD50 of 33,900 mg/kg for rabbits, Fatty (1967) reported an
LD50 of 26,000 mg/kg for mice, and Krauskopf (1973) reported an LD50 of
26,300 mg/kg for guinea pigs. In a study by Dostal et al. (1987), DEHP
at a dose of 100 mg/kg was not lethal to either suckling or weanling
rats, whereas a dose of 1,000 mg/kg was lethal to suckling rats. These
findings indicate that DEHP has a low order of acute oral toxicity, but
that it may be more toxic during early postnatal development. Because
DEHP is a component of a variety of plastic consumer goods, there is a
potential for accidental ingestion. However, no human case studies of
this type were available. This suggests that such incidents are not
common or serious occurrences.

2.2.1.2  Systemic/target organ toxicity
     Animal studies have shown the liver and testes to be the primary
end points for DEHP toxicity.
     Hepatotoxicity (liver).  A number of studies in laboratory animals
have demonstrated that oral exposure to DEHP results in adverse hepatic
effects.  At high oral dose levels, DEHP causes functional hepatic damage
characterized by morphological changes and alterations in the activity
of hepatic enzyme systems.  Several studies have also demonstrated that
DEHP is a peroxisome proliferator, which is of significance because of
an association between peroxisome proliferation and both liver
hyperplasia and liver cancer in laboratory animals. No animal studies
are available on Che hepatic effects of DEHP when administered either by
inhalation or by dermal exposure.

     No human data are available on adverse hepatic effects following
inhalation,  oral, or dermal exposures.  However, Canning et al.  (1984)
reported an increase in the number of peroxisomes in the livers of
patients receiving kidney dialysis treatments.

     Rhodes et al. (1986) reported that oral and intraperitoneal
administration of DEHP to the marmoset monkey at doses up to 5
mmole/kg/day DEHP (equivalent to 1,950 mg/kg/day) for 14 days did not
induce morphological or biochemical changes in the liver comparable to
those obtained in rats administered the same amount of DEHP.

     NTP (1982)  performed single-dose and 14-day repeated-dose oral
studies using F344 rats and B6C3F1 mice.  In the single-dose mouse study.
groups of five animals per sex were treated with various doses (1.25 to
20 g/kg)  of DEHP by gavage.  In the repeated-dose mouse study, groups of
five animals per sex were administered DEHP in the feed (6,300 to
100,000 ppm) for 14 days. No adverse hepatic effects were reported

-------
 14    Section  2

 following  either  single or  repeated doses. These results indicate a
 NOAEL of 20,000 mg/kg  for acute hepatotoxicity in the mouse.

      Lake  et  al.  (1984) treated male Sprague-Dawley rats and Syrian
 hamsters with 25,  100. 250, or 1,000 mg/kg/day DEHP orally for 14 days.
 Liver weights in  the rat were increased significantly at doses of 100
 mg/kg and  greater.  In  the hamster, liver weights were increased
 significantly only at  the 1,000-mg/kg dose. These results indicate a
 marked species difference in this parameter of liver toxicity between
 the rat (NOAEL -  25 mg/kg;  LOAEL - 100 mg/kg) and the hamster (NOAEL -
 250 mg/kg; LOAEL  - 1,000 mg/kg).

      F. E. Mitchell et al.  (1985) evaluated acute and intermediate
 hepatotoxicity in rats fed  DEHP at doses of 50, 200, or 1,000 mg/kg/day
 for 3, 7,  14, or  28 days or 9 months. Histological and biochemical
 alterations were  observed at all doses tested (including 50 mg/kg, the
 LOAEL) for all acute (3, 7, and 14 days) and intermediate (28 days and 9
 months) dosing regimens. Mitchell et al. (1985) regarded the liver
 effects observed  following  the treatment with DEHP to be a compensatory
 phenomenon rather than a pathological event. However, ATSDR considers
 the effects observed in this study to be adverse rather than adaptive.
      Dostal et al.  (1987) investigated the relative sensitivity of
 suckling rats as  compared to adults to the effects of DEHP. Five daily
 oral  doses of 0,  10, 100, 1,000, or 2,000 mg/kg DEHP were administered
 to male Sprague-Dawley rats beginning at 6, 14, 16, 21, 42, and 86 days
 of age. Twenty-four hours after the last dose, rats were sacrificed, and
 plasma cholesterol, triglyceride levels, and the activities of the
 hepatic peroxisomal enzymes, palmitoyl CoA oxidase and carnitine
 acetyltransferase, were determined. Relative liver weight (g/100 g body
 wt) was increased by 30 to  58% at 1,000 mg/kg in all age groups. Smaller
 but significant increases in relative liver weights were also observed
 at doses of 100 mg/kg  (LOAEL) in all but the 1-week-old rats. Absolute
 liver  weight was  increased  at doses of 100 mg/kg or more in all but the
 1-week-old rats.

      In a  study by BIBRA (1987), Cynomolgus monkeys (2 males/group) were
 administered  0, 100, or 500 mg/kg DEHP by gavage for 25 days. There were
 no treatment-related changes in relative liver weight, palmitoyl-CoA
 oxidation,  carnitine acetyltransferase, or lauric acid 11- and 12-hydro-
 zylation.  Histological and  electron-microscopic examination of the
 livers revealed no treatment-related effects.

     Carpenter et al.  (1953) fed groups of 23 to 24 guinea pigs of each
 sex diets containing 0, 400, or 1,300 ppm DEHP for a period of 1 year.
 No treatment-related effects were observed on mortality, body weight,
 kidney weight, or gross pathology and histopathology of the liver. No
 changes in absolute liver weight were observed. However, statistically
 significant increases in relative liver weights were observed in both
 groups of  treated females (19 and 64 ng/kg/day). Carpenter et al. (1953)
 concluded  that despite the  significant increase in liver weights of the
high-dose  females, 64 mg/kg/day was a NOAEL.  However, ATSDR has
determined 19 mg/kg/day to  be a LOAEL. based on the statistically
 significant increases in relative liver weights of both groups of
 treated females.

-------
                                              Health Effects Summary   15

      In a 2-year NTP (1982) bioassay of DEHP,  male F344 rats were fed
 diets which contained 0 (control).  6,000.  or 12.000 ppm DEHP. This was
 equivalent to doses of 0.  322.  and 674 mg/kg/day DEHP.  At the conclusion
 of the study, the rats were sacrificed,  and histological examinations
 were performed.  Hepatic tumors  and peroxisome  proliferation were
 observed at the  high- and  low-dose levels;  therefore,  under the
 conditions of this study the LOAEL was 322  mg/kg/day.

      Jacobson et al.  (1977) evaluated the  effects of DEHP solubilized as
 a result of leaching from  polyvinyl chloride blood bags on the hepatic
 function and histology of  rhesus  monkeys undergoing chronic blood
 transfusions. The average  cumulative amount of DEHP infused over 1 year
 was 0.0584 mg/kg/day. Exposure  to DEHP resulted in abnormal hepatic
 scintillation liver scans  and abnormal sulfobromophthalein (BSP)
 clearance in four of seven animals,  and abnormal morphologic changes  in
 liver histology  in six of  seven animals. Abnormal liver changes included
 vacuolization of Kupffer cells, foci of parenchymal necrosis,  chronic
 inflammatory cell infiltrates,  and binucleate  cells.

      Testicular  toxicity.   DEHP has  also been  shown to  induce testicular
 toxicity in numerous  animal studies.  Such effects,  however,  are
 generally considered  as  evidence  of  reproductive toxicity and are
 therefore discussed in detail in  the section on reproductive toxicity
 (Sect.  2.2.1.4).

 2.2.1.3  Developmental  toxicity

      A number of studies in laboratory animals  have demonstrated that
 oral  exposure to DEHP induces developmental  toxicity. No animal studies
 are available on the  developmental  toxicity  of  DEHP when administered
 either by inhalation  or  by  dermal exposure.  No  human data are  available
 on  adverse developmental effects  following  inhalation,  oral,  or dermal
 exposures.

      Nakamura et al.  (1979)  evaluated teratogenicity in wy-Slc x CBA
 mice  administered a single  oral dose  of  0.05, 0.1,  1.0,  2.5,  5.0,  or
 10.0  raL/kg DEHP  on day  7 of gestation. Based on a density of 0.9861 for
 DEHP  (IARC 1982),  these  doses were equivalent to 49.3,  98.6,  986,  2,455,
 4,930,  and 9,860  mg/kg.  DEHP  resulted in a decrease in  body  weight of
 live  fetuses  at  doses of 49.3 mg/kg  (the LOAEL)  and higher.  At doses  of
 98.6  mg/kg and higher, DEHP also resulted in deformed or dead  fetuses.

      Wolkowski-Tyl et al.  (1984a) fed timed  pregnant Fischer 344 rats
 nominal  doses of  0, 5,000,   10.000, 15,000,  or 20,000 ppm DEHP  in the
 diet  on  days  0 to  20  of  gestation. Based on  food consumption data,  this
 was equivalent to  doses  of  approximately 0,  357.  666. 856, and 1.055
 mgAg/day,  respectively. Maternal toxicity,  as  evidenced by
 significantly reduced body weight gains  in the  dams  receiving  10,000.
 15.000,  and 20,000 ppm of DEHP,  was observed. The number and percentage
 of  resorptions, nonlive  fetuses, and  nonlive malformed  fetuses were
 increased  in a dose-related manner, with a statistically significant
 increase in all three parameters in the high-dose group  when compared
with  the controls. The number of live  fetuses per litter exhibited a
dose-related decrease, with the  high-dose group  significantly  lower than
 the controls. Among the  live neonates, there was  a  dose-related decrease
 in  the body weights of both sexes, with a statistically  significant

-------
16   Section 2

decrease in fetal weight at all doses tested. There was also a
significant dose-related trend in the percentage of malformed fetuses
per litter. However, there were no significant pairwise comparisons in
the number of fetuses malformed per litter. Under the conditions of this
study, DEHP produced significant developmental toxicity, exhibited as
decreased fetal weight, at doses of 10,000, 15,000, and 20,000 ppm.
ATSDR has concluded that the LOAEL was 5,000 ppm (357 mg/kg/day) and
that a NOAEL could not be determined from this study.

     Wolkowski-Tyl et al. (1984b) reported that DEHP was developmentally
toxic to CD-I mice exposed during the entire gestational period (days 0
through 17) at dose levels that produced significant maternal and other
fetal toxicity. The mice received diets containing 0, 250, 500, 1,000,
or 1,500 ppm DEHP. Based on food consumption data, this was equivalent
to approximately 0, 44, 91, 191, or 292 mg/kg/day. Major malformations
included external, visceral, and skeletal effects, observed at doses of
91 mg/kg/day (the LOAEL) and higher. Under the conditions of this study
the NOAEL was determined to be 250 ppm (44 mg/kg/day) DEHP in the diet,
if no significant maternal or fetal toxicity had been observed.

2.2.1.4  Reproductive toxicity

     A number of studies in laboratory animals have demonstrated that
oral exposure to DEHP induces adverse testicular effects, characterized
by decreased organ weights and histological changes in the seminiferous
tubules. Hono(2-ethylhexyl)phthalate (HEHP), a monoester metabolite
formed in the gastrointestinal tract, has also been reported to produce
testicular damage (Oishi and Hiraga 1980).  No animal studies are
available on the testicular effects of DEHP when administered either by
inhalation or by dermal exposure. No human data are available on adverse
testicular effects following inhalation, oral, or dermal exposures.
     Persons who may be at risk from reproductive and developmental
toxicity from DEHP exposure are individuals receiving hemodialysis
treatments. DEHP is frequently used as a plasticizer for medical vinyl
products, such as blood bags, intravenous administration sets, and
hemodialysis tubing. Trebbin (1979) discussed four case studies in which
pregnant women with chronic renal failure underwent hemodialysis. These
women successfully conceived and concluded the pregnancy with the birth
of apparently healthy, normal infants. Bailey (1977) discussed the
effect of hemodialysis on men with chronic renal failure. Prior to
treatment,  the testicles become soft and atrophic, spermatogenesis
ceases, some Leydig cells are lost, plasma testosterone levels decrease,
and libido is lost. Hemodialysis resulted in restoration of their sex
drive and spermatogenesis. Conception and completely successful
pregnancies in their spouses followed. This suggests that intravenous
exposure to DEHP--if this was the plasticizer involved in these cases--
may not pose significant risks of reproductive toxicity in male dialysis
patients.

     Sjoberg et al. (1985c) evaluated the testicular toxicity of 1.0
g/kg/day DEHP in 25-, 40-, and 60-day-old rats. Following 14 daily oral
doses, testicular damage was observed in the 25-day-old group but not  in
the 40- or the 60-day-old group. These findings suggest an age-dependent
susceptibility to the testicular effects of DEHP. However, in a follow-
up study by Sjoberg et al. (1985b), age-dependent testicular effects

-------
                                             Heal eh Effects Summary   17

were not evident following intravenous DEHP administration. This is
likely because of the differences  in the route of administration.  DEHP
is extensively converted to MEHP in the gastrointestinal tract  HEHP
would not be formed to the same extent after intravenous administration.

     Rhodes et al.  (1986) evaluated the testicular toxicity of DEHP Ln
marmoset monkeys administered (orally or intraperitoneally) doses up to
5 mmole/kg/day (1,950 mg/kg/day).  Significant biochemical and
morphological changes were not evident, even at the highest dose tested
(1,950 mg/kg/day, the NOAEL).
     Aearwal et al. (1986) evaluated the reproductive (testicular)
toxicity of DEHP in sexually mature male F344 rats. DEHP was
administered in the diet at levels of 0, 320  1.250  5.000. o,: 20 000
ppm  (equivalent to 0. 16, 62.4. 250. or 1,000 ^As/day) for 60 days.
Reduced body weight, testicular weight, and epididymal weight were
observed at doset of 250 mg/kg/day  (the LOAEL) and higher  but not at
doses of 62.5 mgAs/day  (the NOAEL) and lower. A decrease  in various sex
hormone levels was also  observed at the two highest DEHP levels, with
degenerative testicular  alterations and reduced epidiaymal sperm count
and  motility noted only  at  the highest level.
      In a  study performed by Reel et  al.  (1982), groups of mice were  fed
0  100, 1.000. or  3.000  ppm DEHP in the diet  during a 7-day premating
oeriod and a 98-day cohabitation period. These dose levels are
equivalent to approximately 0,  13,  130, and  390  mg/kg/day. Fertility  was
completely suppressed  in the high-dose group  and significantly
 (P < 0 05) reduced in  the  intermediate-dose  group.  DEHP was a
 reproductive  toxicant  in male  and  female  CD-I mice, as  evidenced by a
decreased  fertility  index,  decreased  number  of litters   decreased number
 of live pups, and  a  lower  proportion  of live  pups  per  litter. «"*««"•
 conditions of this study,  the  NOAEL was determined to  be  13 mg/kg/day.
 and the LOAEL was  determined  to be  130 mg/kg/day.
      In a  2-year NTP (1982) bioassay  of DEHP (previously discussed).
 male F344  rats  were fed diets  which contained 0 (control), 6,000,  or
 12 000 ppm DEHP. This  was equivalent  to doses of 0.  322,  and &74
 mg/kg/day  DEHP.  At the conclusion of  the  study the rats were sacrificed.
 and historical examination revealed seminiferous tubule degeneration
 of the testes in 1/43 controls (2.3%). 2/44 low-dose males (4.5%)  and
 43/48 high-dose males (89.6%). Therefore, under the conditions of this
 study the LOAEL was 674 mg/kg/day and the NOAEL was 322 mg/kg/day.

 2.2.1.5  Genotoxlcity
      DEHP has been tested extensively in short-term genotoxicity  assays
 because of its reported carcinogenicity and  its Inclusion in the
 collaborative study program of the International Program  on Chemical
 Safety  DEHP has not been  shown to be mutagenic in most microbial and
 mammalian assay systems. In addition, the majority of  the data also
  suggest that MEHP and 2-ethylhexanol. the major metabolites  of DEHP.  are
 not mutagenic.

-------
 18   Section 2

 2.2.1.6  CareInogenic ity

      There were  no available  data  on  the potential carcinogeniclty of
 DEHP in humans exposed via  inhalation, oral, or dermal exposures.
 Neither were data available on  inhalation or dermal exposures  in
 experimental animals.

      The carcinogenicity of DEHP has  been tested in a bioassay using
 Fischer 344 rats and B6C3F1 mice (NTP 1982, Kluwe et al. 1982a,b).
 Croups  of 50 rats per  sex were  fed diets which contained 0  (control),
 6.000,  or 12,000 ppm DEHP,  and  groups of 50 mice per sex were  fed diets
 which contained  0 (control),  3,000, or 6,000 ppm of DEHP (purity 95.5%)
 for  103 weeks. These dose levels were equivalent to 0, 322, and 674
 mg/kg/day for the male rats;  0, 394,  and 774 mgAg/day for  the female
 rats; 0,  672,  and 1,325 mgAg/day  for the male mice; and 0, 799. and
 1,821 mgAg/day  for the female  mice,  respectively. A 1- to  2-week
 nontreatment observation period was allowed after the termination of
 exposure,  after  which  all survivors were sacrificed and examined both
 grossly and microscopically.  Histopathological examinations were
 conducted on all animals sacrificed or discovered dead (if  not precluded
 by autolysis).

      A  summary of the  incidence of hepatocellular carcinomas and
 neoplastic nodules  observed in  the rats is presented below:

                          Dose
                         (ppm)    Males   Females
                             0    3/50     0/50
                         6,000    6/49     6/49
                        12,000   12/49    13/50

     In female rats, the incidence of hepatocellular carcinomas at the
higher dose was greater than that in controls by pairwise comparisons,
and there was a significant (P < 0.05) dose-related trend. A dose-
related trend was also observed for hepatocellular carcinomas in the
males, but pairwise comparisons did not indicate a statistically
significant increase.

     A summary of the incidence of hepatocellular carcinomas and
neoplastic nodules observed in the mice is presented below:

                        Dose
                        (ppm)   Males   Females
                            0   14/50     1/50
                        3,000   25/48    12/50
                        6,000   29/50    18/50
     In mice, there was a statistically significant increase in the
incidence of hepatocellular carcinoma in both the high-dose (P - 0.022)
males and high-dose females (P < 0.001) when compared to the controls. A
significant (P < 0.05) dose-related trend of increasing hepatocellular

-------
                                             Health Effaces Summary   19

 carcinoma was  also  observed  for  Che mice of both  sexes. Metastases of
 the hepatocellular  carcinomas were observed in  the lungs of 12  treated
 males and 8  treated females.

      Northrup  et  al.  (1982)  have questioned the validity of the NTP
 bioassay of  DEHP. suggesting that the maximum tolerated dose  (MTD) was
 exceeded in  both  species because body weight gain was  depressed by more
 than 10% in  several of  the treatment groups. However,  Kluwe et al.
 (1983)  have  pointed out that the 10% weight differential is only a
 guideline and  that  the  primary reason for not exceeding the estimated
 HTD is  to avoid excessive early mortality which might  prevent tumor
 development, and  to avoid pathological lesions other than neoplasia. In
 this study,  survival  was not adversely affected by DEHP, and  statistical
 analyses failed to  demonstrate any correlation between the occurrence of
 nonneoplastic  tissue  lesions (testicular atrophy  in the high-dose male
 rats and mice  and pituitary  hypertrophy in the high-dose male rats) and
 the development of  hepatocellular tumors in rats  and mice of  both sexes
 (Kluwe  et al.  1982a,b 1983).

      Northrup  et  al.  (1982)  also questioned the significance  of the
 DEHP-induced increase in liver tumors in the NTP  bioassay because of
 intralaboratory variations in incidences of liver tumors in controls in
 bioassays conducted simultaneously in the same rooms as the DEHP study.
 They reported  that  statistical analysis indicated that there was a
 significantly  lower incidence of tumor-bearing animals among  the female
 mice used in the  DEHP control group.-In addition, they found  that the
 number  of tumor-bearing male or female mice in the DEHP treatment groups
 did not substantially differ from the average for all pooled  controls.
 They also found that  in male rats,  the incidence  of tumor-bearing
 animals had  a highly  negative dose response (? <  0.001), and  female racs
 had a marginally  positive dose response (? - 0.045) when paired to the
 pooled  controls. However,  Kluwe et al.  (1983) reported that there was
 relatively little variation  in liver tumor incidences among NTP
 historical controls,  and that the data reported in the concurrent study
 controls  were quite similar  to the historical controls. In addition,
 Kluwe et  al.  (19B2a,b) have  reported that replacement of the concurrent
 study controls with concurrent laboratory controls or pooled controls
 from bioassays performed at approximately the same time did not alter
 the  statistical significance of the DEHP effects. Replacement of the
 concurrent study controls with historical controls strengthened the
 statistical  significance of  the difference in liver tumor incidence.

     In a draft document,  the EPA (1986a) has classified DEHP as a
 probable  human carcinogen,  B2,  according to the criteria in the EPA
 Guidelines for Carcinogen Risk Assessment (EPA 1986a).  Evidence on
 potential carcinogenicity from animal studies is  "Sufficient," and
 evidence  from human studies is "No  Data" (EPA 1986a).

     Various  types of mathematical  extrapolation  models can be fit to
 the NTP carcinogenesis bioassay data of DEHP in F344 rats and B6C3F1
mice.   Different high-to-low dose extrapolation methods may give a
 reasonable fit to the observed data but may lead  to differences in the
estimates of  risk at low doses.  The choice of the model depends largely
on the assumed mechanism of action   If  DEHP is a  genotoxic carcinogen,
 it is capable of direct reaction with genetic material. Under these

-------
20   Section 2

circumstances, the dose-response curve  is  expected to be linear at low
doses. However, if DEHP acts by an epigenetic mechanism, the dose-
response curve may be subject to individual  tolerances,  with each
individual having a specific threshold  below which no effect would
occur, and the aggregation of these individual tolerances would fit a
specific statistical distribution for the  general population.  The issue
of whether DEHP acts by a genotoxic or  nongenotoxic mechanism has not
been resolved at the present time. In keeping with the EPA Risk
Assessment Guidelines (EPA 1986) and the OSTP Cancer Principles (OSTP
1985), the choice of the low-dose extrapolation method is governed by
consistency with current understanding of  the mechanism of
carcinogenesis and not solely on goodness-of-fit to the observed data.
When data are limited, and when uncertainty  exists regarding the
mechanisms of carcinogenic action, the  OSTP  Principles suggest that
models or procedures which incorporate  low-dose linearity are preferred
when compatible with the limited information available.  The EPA
Guidelines recommend that the linearized multistage procedure be used in
the absence of adequate information to the contrary.

     In evaluating the potential carcinogenicity of DEHP by the EPA (EPA
1987c), slope factors were estimated by fitting the liver tumor data in
male and female rats and mice with the linearized multistage model. The
unit risk value is estimated to be 4.0 x 10"? for drinking water
containing 1 pg/L DEHP. The drinking water concentrations of DEHP
corresponding to lifetime excess cancer risks of 10'4,  10'5, and  10'6
are 0.3, 0.03, and 0.003 mg/L, respectively.

     Computations of the unit risk for the experimental animals were
made by the Consumer Product Safety Commission (CPSC) (1985) assuming
the generalized multistage model of carcinogenesis. Risk estimates were
performed using the Global 83 computer program of Howe  and Crump  in its
unrestricted  form. The lifetime excess risk to the experimental animals
per mg/kg/day for hepatocellular carcinomas and neoplastic nodules
following exposure to DEHP was estimated by CPSC (1985) as follows:

                     Lifetime animal risk estimates
                             [(mg/kg/day)-1]
Species
Rat
Mouse
Sex
Male
Female
Male
Female
Maximum
likelihood
estimate
0.000154
0.000285
0.000678
0.000545
Upper 95%
confidence
limit
0.000471
0.000523
0.001122
0.000752
     These estimated  animal  risks can be converted  to human  risk
estimates by  applying animal-to-human extrapolation factors  that are
based on comparative  body  surface areas of humans vs rats  and mice.
These extrapolation factors  are  5.6  for rats  and 12.8 for  mice.  The
resulting human  risk  extrapolations  are:

-------
                                              Health  Effaces Summary   21
                       Lifetime human risk estimates
                              «nig/kg/day)-1]
Animal on which based

Species
Rat

Mouse


Sex
Male
Female
Male
Female
Maximum
likelihood
estimate
0.000862
0.00160
0.00868
0.00698
Upper 95%
£ • 1
limit
0.00264
0.00293
0.0144
0.00963
      Note that the most conservative (highest) risk estimate derived by
 CPSC (1985) of 0.0144 is virtually identical to the 0.015 potency factor
 derived by EPA (1986b).  However,  EPA based its risk estimates on
 hepatocellular carcinomas in female rats,  whereas the highest risk
 estimate reported by CPSC (1985)  is based on liver tumors in the male
 mouse.

      Because of what appeared to  be excessive reported food consumption
 by the  mice (8.1 to 8.4  g/day),  the CPSC (1985) converted the dose data
 for mice to a milligram  per kilogram basis using an estimated average
 daily consumption of 3.9 g per  animal.  The CPSC's rationale for this was
 that the food fed the mice was  not in pelleted form,  and some of the
 reported food consumption was due to substantial scattering.  The risk
 estimate performed by EPA did not incorporate an adjustment for food
 consumption;  therefore,  the lifetime extra carcinogenic risk may be
 underestimated.

 2.2.2   Biological Monitoring as a Measure  of  Exposure  and Effects

     As discussed in Sect.  4, Toxicological Data,  DEHP (or its
 metabolites)  is well absorbed in  the  gastrointestinal  tract following
 oral exposure. Once  absorbed, it  is  widely distributed in the body,  with
 the  liver being the  major  repository  organ. As  discussed in the  toxicity
 review  of Sect. 4,  the major  target  organs of DEHP  appear to  be  the
 liver and testes.

     DEHP and  its metabolites. MEHP and 2-ethylhexanol.  can be measured
 in urine and blood  in order  to confirm prior recent exposure. Monitoring
 urine for DEHP will  not be a  good marker for prior  DEHP exposure  because
 of poor solubility.  There  is  scant information on the  relationship
 between DEHP uptake  and its elimination in the urine in humans. At
 present, there are no data available  for assessing  the  potential
 correlation between  the values obtained with these measurements and  the
 toxic effects observed in experimental animals. Also, because DEHP  is
 rapidly  metabolized and excreted,  these  methods are not  good  indicators
of previous exposures. In addition, because MEHP and 2-ethylhexanol
undergo  extensive metabolism, they are also poor markers  of previous
exposure.

-------
22   Section 2

2.2.3  Environmental Levels as Indicators  of  Exposure  and Effects

2.2.3.1  Levels found in the environment
     There are no available case studies or epidemiological
investigations that suggest that levels of DEHP  found  in the
environmental media are associated with significant  human exposure  or
adverse health effects.

2.2.3.2  Human exposure potential
     No data have been located to suggest  that there are chemical-
specific issues relevant to estimating the body-dose,  tissue levels,  or
health effects from DEHP concentrations in soil,  water,  or food.

2.3  ADEQUACY OF DATABASE

2.3.1  Introduction
     Section 110 (3) of SARA directs the Administrator of ATSDR to
prepare a toxicological profile for each of  the  100  most significant
hazardous substances found at facilities on  the  CERCLA National
Priorities List. Each profile must include the following content:
    "(A)  An examination, summary, and interpretation  of available
          toxicological information and epidemiologic  evaluations on a
          hazardous substance in order to  ascertain  the levels of
          significant human exposure for  the  substance and the
          associated acute, subacute, and  chronic health effects.
     (B)  A determination of whether adequate information on the health
          effects of each substance is available or  in the process  of
          development to determine levels  of exposure  which present a
          significant risk to human health of acute, subacute, and
          chronic health effects.
     (C)  Where appropriate, an identification of toxicological testing
          needed to identify the types or  levels of exposure that may
          present significant risk of adverse health effects in humans."

     This section identifies gaps in current knowledge relevant to
developing levels of significant exposure  for DEHP.  Such gaps are
identified for certain health effect end points (lethality,
systemic/target organ toxicity, developmental toxicity, reproductive
toxicity, and carcinogenicity) reviewed in Sect. 2.2 of this profile in
developing levels of significant exposure  for DEHP,  and for other areas
such as human biological monitoring and mechanisms  of toxicity. The
present section briefly summarizes the availability of existing human
and animal data, identifies data gaps, and summarizes research in
progress that may fill such gaps.
     Specific research programs for obtaining data  needed to develop
levels of significant exposure for DEHP will be developed by ATSDR.  NTP,
and EPA in the future.

-------
                                              Health Effects Summary   23

 2.3.2  Health Effect End Points

 2.3.2.1  Introduction and graphic  summary

      The availability of data  for  health  effects  in humans and animals
 is depicted on bar graphs In Figs.  2.5  and 2.6,  respectively.

      The bars of full height indicate  that there  are data to meet  at
 least one of the following criteria:

  1.   For noncancer health end  points, one or  more studies are available
      that meet current scientific  standards and are sufficient to  define
      a range of toxicity from  no-effect levels  (NOAELs)  to levels  that
      cause effects (LOAELs or  FELs).

  2.   For human carcinogenicity, a  substance is classified as either a
      "known human carcinogen"  or a  "probable  human carcinogen" by  both
      EPA and IARC (qualitative), and the  data are sufficient to derive a
      cancer potency factor (quantitative).

  3.   For animal carcinogenicity, a  substance  causes a statistically
      significant number  of tumors  in at least one species,  and the  data
      are sufficient to derive  a cancer  potency factor.

  4.   There are  studies which show  that  the chemical does  not cause  this
      health effect via this  exposure route.

      Bars  of half height  indicate that  "some" information for  the  end
point  exists, but does not meet any of  these  criteria.

      The absence  of a  column indicates  that no information exists  for
that  end point  and route.

2.3.2.2  Description of highlights of graphs

     As  shown in  Figure 2.5, there were no adequate human data for  DEHP.
There  were  no available case studies or epidemiological reports
providing  information  on exposure levels  associated with  any reported
effects.

      In the  data based on animal studies,  as  shown  in  Figure 2.6,  there
were no data available via the  dermal route. The  few studies conducted
via inhalation were not sufficient to assess  the  effects
(lethality/decreased longevity) that were being tested. Studies using
the oral route of administration were adequate for  acute,  intermediate,
and chronic exposures  to DEHP.  with the liver being the most sensitive
target organ. The NTP carcinogenicity bioassay was  also adequate for
assessing carcinogenicity. Data on lethality,  developmental  toxicity.
and reproductive toxicity following oral administration were considered
adequate.

2.3.2.3  Summary of relevant ongoing research

     A search of the NIH CRISP  (Computerized Retrieval of  Information on
Scientific Projects) database revealed that several  investigations  of
the role of peroxisomal proliferation in DEHP-induced carcinogenicity

-------
                                           HUMAN DATA
                                                                                                ORAL
                                                                                                                           01
                                                                                                                           r>
                                                                                                                           rt
                                                                                                                           >-.

                                                                                                                           §
                                                                                                      J
                                                                                                         ^ SUFFICIENT

                                                                                                          INFORMATION*
                                                                                                      J
                                                                                                          ^   SOME

                                                                                                          INFORMATION
                                                                                                               NO

                                                                                                          INFORMATION
                                                                                            INHALATION
                                                                                       DERMAL
LETHALITY       ACUTE     INTERMEDIATE    CHRONIC   DEVELOPMENTAL REPRODUCTIVE  CARCINOOEMCITV

           Z	/    TOXICITY       TOXICITY
                    SYSTEMIC TOXICITY



                     'Sufficient information exists to meet at least one of the criteria for cancer or noncancer end points.



                 Fig. 2.5. Availability of information on health effects of d«2-«lhylhexyl)phih«late (human data).

-------
                                           ANIMAL  DATA
                                                                                                           V  SUFFICIENT

                                                                                                           'INFORMATION*
                                                                                                           V    SOME

                                                                                                           'INFORMATION
                                                                                                                  NO

                                                                                                             INFORMATION
                                                                                              INHALATION
                                                                                         DERMAL
LETHALITY
                ACUTE
                         INTERMEDIATE
                                               -

CHRONIC   DEVELOPMENTAL  REPRODUCTIVE  CAHCINOOENICITY


            TOXICITT       TOXICITV
                                                                                                                              Cki
                                                                                                                              t-4
                                                                                                                              n
(b
n
rt
to
                     SYSTEMIC TOXICITY



                     'Sufficient information exists to meet at least one of the criteria for cancer or noncancer end points.



                  Fig. 2.6. Availability of information on health effects of di(2-etnylhexyl)phthalate (animal data).

-------
26   Section 2

are being conducted by researchers at the National Institutes of Health.
These studies are summarized in Table 2.1.  Investigations of DEHP are
being conducted by other research groups such as the Chemical Industry
Institute on Toxicology (CUT). In addition,  a bioassay for the
metabolite 2-ethylhexanol is being planned by the National Toxicology
Program (NTP).

2.3.3  Other Information Needed for Human Health Assessment

2.3.3.1  Fharmacokinetics and mechanisms of action

     The mechanism of DEHP-induced careinogenieity is currently not well
understood. Most of the evidence from genotoxicity assays indicates that
DEHP is nongenotoxic. DEHP appears to belong to a class of carcinogens
that induces proliferation of peroxisomes and liver cells. A
hypothetical, but unproven, mechanism for DEHP-induced carcinogenesis is
that peroxisome proliferation causes an imbalance in reactive oxygen
species that somehow initiates the carcinogenic process. In the NTP
bioassay, excess cancers were found only in the liver, the site of the
greatest peroxisome activity.
     Additional research is necessary to elucidate the exact mechanism
of action responsible for the carcinogenic effects of DEHP.
     A second area currently being investigated is that of the role of
zinc in the testicular effects of DEHP. DEHP-induced testicular injury
is accompanied by a decrease in the zinc content of the testes and an
increase in the urinary excretion of this element. At present, the
significance of these findings is unclear.  Further research is necessary
to elucidate the role of DEHP in testicular effects.
     Additional metabolic and pharmacokinetic data are needed for the
purpose of interspecies and high-to-low dose extrapolation of the toxic
effects of DEHP.

2.3.3.2  Monitoring of human biological samples
     There are two methods to test for human exposure to DEHP. These
methods involve analysis of blood or urine for the presence of DEHP
and/or its major metabolites, HEHP and 2-ethylhexanol. Because DEHP is
rapidly metabolized and excreted (and apparently does not
bioaccumulate), these methods are appropriate only for measuring recent,
prior exposure.

2.3.3.3  Environmental considerations

     Current methodologies for assessing the levels of DEHP in the
environment are adequate. DEHP is relatively nonvolatile at ambient
temperatures, so that partitioning into the atmosphere is extremely
limited. Because DEHP is readily degraded under aerobic conditions,
environmental accumulation of levels toxic to humans  is not expected.
This has been confirmed by environmental monitoring and analytical
studies conducted under controlled conditions.

-------
                                           Health  Effects Summary   27
              Table 2.1. Summary of ongoing research"
Principal
investigator
Melnick, R. L.
Institutional
affiliation
NIEHS
Description
of research
Investigate role
of peroxisomal
production in
DEHP-induced
hepatotoxicity
Data
deficiency
Mechanism of
DEHP-induced
hepatotoxicity
Melnick, R. L.   NIEHS
Determine the role
of zinc in the
pathophysiology of
DEHP-induced
reproductive
effects
Mechanism of
DEHP-induced
reproductive
toxicity
Ward, J. M. NCI


Diwan, B. NCI


Investigate tumor
initiation and
promotion by DEHP
Investigate tumor
promotion by DEHP

Mechanism of
DEHP-induced
carcinogemcity
Mechanism of
DEHP-induced
carcinogemcity
   "Adapted from NIH CRISP database.

-------
                                                                      29
                 3.  CHEMICAL AND PHYSICAL INFORMATION

3.1  CHEMICAL IDENTITY

     The chemical formula,  structure,  synonyms, trade names, and
identification numbers for DEHP are listed in Table 3.1.

3.2  CHEMICAL AND PHYSICAL PROPERTIES

     Important physical and chemical properties of DEHP are listed in
Table 3.2.

-------
30
Section  3
                           T.Me 3.1.  Chemical identity of dK2-etbylbexyl)pfathalaMa
          Chemical name

          Synonyms
          Trade name*
          Chemical formula

          Wuwesser line notation
          Chemical structure
                                        Di(2-ethylhexyl) phthalate

                                        1.2-Bcnzenedicarboxylic acid, bis(2-ethylhexyl) ester
                                        Phthahc acid. b»(2-ethylhexyl) ester
                                        BEHP
                                        1.2-Benzenedicarboxylic and. bis(ethylhexyl) ester
                                        But 2-ethylhexyl )- 1 ,2-benzenedicarboxylate
                                        Bis(2-ethylhexyl) phthalate
                                        DEHP
                                        Di(2-ethylbexyl) orthophthalate
                                        Di(ethylhexyl) phthalate
                                        Dioctyl phthalate
                                        OOP
                                        2-Eihylhexyl phthalate
                                        Octyl  phthalate
                                        Phthalic acid, dioctyl ester

                                        Bisoflex 81. Bisoflex OOP, Compound 889.
                                        DAF 68. Ergoplast FDO, Evtplast 80.
                                        Eviplast 81. Flexunel. Flexol OOP.
                                        Good-Rite GP 264. Hatcol OOP. Kodaflex OOP,
                                        Mollan O. Nuoplaz OOP, Octoil.
                                        Palatinol AH. Palatinol OOP. Pittsburg PX-138.
                                        Reomol OOP. Reomol D 79P. Sicol ISO.
                                        Staflex OOP, Truflex OOP. Vestmol AH.
                                        Vmicizer 80. Witcizer 312
                                        4Y2&1OVRBV01Y4&2
                                                               COOCH2CH(C2H8)(CH2)!,CH3
          Identification numbers
            CAS Registry No.                     117-81 -7
            NIOSH RTECS No.                 TI0350000
            EPA Hazardous Waste No.            UO28
            OHM-TADS No.                    7216693
            DOT/UN/NA/IMCO Shipping No    NAI693
            STCC No.
            Hazardous Substances Data Bank No   339
            National Cancer Institute No.          CS2733
              'References.  HSDB 1987. I ARC 1982

-------
                           Chemical  and  Physical  Information    31
  Tabk 3.2. Physical and chemical properties of dK2-etbylbexyl)pfetkalate
Property
Molecular weight
Color
Physical state
Odor
Odor threshold
Melting pout

Boiling point
Autoignition temperature
Solubility
Water


Organic solvents



Density
Vapor density

Partition coefficients
Octanol-water (log !(„)
Soil-organic carbon-water
<*.), mL/g
Vapor pressure
At 200°C
At 25°C
Henry's law constant
Refractive index
Flash point (open cup)
Flammable limit
Conversion factor

Value
39054
Colorless
Liquid
Slight
NDa
-50°C

385°C at 760 mm Hg
390-C

0 285 mg/L at 24° C
0 40 mg/L at 20°C
0 34 mg/L at 2S°C
Miscible with mineral
oil and hexane;
soluble in most
organic solvents
09861 al20°C
160 (air - 1)


488

ND"

1 32 mm Hg
34 X I0~~mm Hg
1 X 10~4atm-mJ/mol
1 4836 at 20° C
215°C
0 3% at 245°C
1 ppm — 1 5 94 mg/m

References
ACCIH 1986
CHRIS 1978
IARC 1982
CHRIS 1978

Clayton and
Clayton 1981
Verschueren 1983*
NFPA 1978*

Verschueren 1983*
Branson 1980
CMA 1983


ACGIH 1986

IARC 1982
Clayton and
Clayton 1981

b



Fnssel 1956

Wolfe et al 1980*
IARC 1982
NFPA 1978*
NFPA 1978*
Clayton and
Clayton 1981
aND - no data.
*Cited in HSDB 1987.

Source  Adapted from HSDB 1987

-------
                                                                       33
                          4.   TOXICOLOGICAL DATA

 4.1   OVERVIEW

      Phchalate  esters  such as  DEHP  are well  absorbed from  che
 gastrointestinal  tract following oral administration.  Hydrolysis  to  the
 corresponding monoester metabolite. MEHP.  with  release of  an alcoholic
 substituent,  2-ethylhexanol,  largely occurs  prior  to intestinal
 absorption. Once  absorbed, DEHP and MEHP are widely  distributed in the
 body,  with  the  liver being the major, initial repository organ.
 Clearance from  the body is rapid, and there  is  only  a  slight cumulative
 potential.  DEHP is converted principally to  polar  derivatives of  the
 monoesters  by oxidative metabolism  prior to  excretion.  In  general, there
 are only quantitative  differences in the metabolite  profiles of phase 1
 oxidations  between species. It had  been previously thought that none of
 the five major  metabolites in rats, all of which involve oxidation of
 methyl groups to  carboxylic acid groups, occurred  in man.  Recently,
 however, it has been shown that one of the two  major metabolites  in man
 involves oxidation of  a terminal methyl group to the corresponding acid.
 However, there  appears  to be major  interspecies differences in the
 phase  2 metabolism of  DEHP. Primates (man, African green monkey,  and
 marmoset) glucuronidate DEHP at the carboxylate moiety following
 hydrolysis of a single ester linkage. Rats can  glucuronidate MEHP, but
 high 0-glucuronidase activity apparently converts  this  phase 2 conjugate
 back to MEHP. Mice appear to be unable to glucuronidate the monoester
 metabolite and, thus, oxidize the residual alkyl chain instead to
 various hydroxy, ketone, and carboxylate derivatives.  The  major route of
 DEHP elimination from  the body is urinary excretion. The relationship of
 DEHP pharmacokinetics  to its toxicological actions is  unknown at  the
 present time, largely due to a lack of elucidated  mechanisms of toxic
 action.

     A number of studies have been conducted to investigate the acute
 toxic effects of DEHP.  When administered by  the oral,  intraperitoneal,
 intravenous, and inhalation routes.  DEHP has a  low order of acute
 toxicity.

     The target organs for DEHP appear to be the liver and testes. DEHP
has been found to induce morphological and biochemical changes in the
 liver of exposed rodents at relatively high dose levels. Similar effects
have been reported for a number of chemicals which induce  hepatic
xenobiotic metabolizing capabilities. The testicular effects of DEHP are
characterized by a decrease in relative organ weight and damage to the
seminiferous tubules.  Similar effects have been reported in animals
treated with MEHP, a major metabolite of DEHP.

     Studies in rats and mice suggest that DEHP is developmentally
toxic.  In the rat, a variety  of congenital abnormalities have been

-------
 34   Section 4

 observed In the offspring of DEHP-treated  dams.  In the  mouse,  the
 developing nervous system appears  to  be  the major  target  site, producing
 exencephaly and splna blflda.  DEHP  Is a  reproductive  toxicant  In male
 and female mice,  resulting In reduced fertility  and both  production of
 fewer litters by breeding pairs  and decreased  litter  size.

      A large database exists on  the genotoxicity of DEHP. DEHP has been
 subjected to extensive testing in bacterial and  both  In vitro and in
 vivo mammalian assay systems.  The weight of the  evidence  suggests that
 DEHP is not mutagenic in bacterial  or mammalian  test  systems.

      In a study by NTP,  DEHP induced  an  increase in the Incidence of
 hepatocellular tumors in both sexes of Fischer 344  rats and B6C3F1 mice
 There was a trend toward increasing numbers of tumors with Increasing
 doses.  In a draft report,  the  EPA has concluded  that  DEHP is a probable
 human carcinogen classified as weight-of-evidence Group B2.

 4.2   TOXICOKINETICS

 4.2.1  Absorption

      Studies of the absorption of DEHP following an oral dose Indicate
 that the  parent compound is initially hydrolyzed by a nonspecific lipase
 in  the  gastrointestinal  tract  to produce mono(ethyIhexyl) phthalate
 (MEHP)  and 2-ethylhexanol.  MEHP  is  readily absorbed from the
 gastrointestinal  tract.  There  is evidence for very  low  level absorption
 of DEHP through the skin.  Although  quantitative  information is not
 available,  animal studies  indicate  DEHP is absorbed by  the lungs.

 4.2.1.1  Inhalation

      Human.   No data are available  on the absorption of DEHP by humans
 exposed via inhalation.

      Animal.   No  quantitative  data  are available on the absorption of
 DEHP by animals exposed  via inhalation. However, acute  inhalation
 toxicity  studies  with rats  performed  by the WARF Institute (1976) and
 Pegg (1979)  have  demonstrated  that  DEHP is absorbed by  the lung. In a
 chronic inhalation study,  Schmezer  et  al. (1988) have shown absorption
 of saturated vapors of DEHP by hamsters.

 4.2.1.2  Oral

      Human.   Information on the oral  absorption  of  DEHP in humans is
 limited. Shaffer  et al.  (1945) reported that a single oral dose of
 10,000  mg of DEHP by a human subject  was recovered  as an unspecified
 phthalate equivalent in  the urine after 24 h. Approximately 4.5% of the
 administered dose was  recovered.  Schmid and Schlatter (1985) found that
 DEHP  taken  orally by two volunteers (30 mg each) was excreted as
 derivatives  of MEPH in the  urine at 11 and 15% of the dose. DEHP taken
by the  same  volunteers for  4 days at  10 mg daily gave no evidence of
 accumulation;  15  and 25% of the total  dose was recovered in the urine.
No information  of fecal  recoveries  of  the parent compound or its
metabolites  was reported.

-------
                                                 Tax Leo Logics I Data   35

     Animal.  DEHP appears to be efficiently absorbed  from  the
 gastrointestinal tract of the rat. Williams and Blanchfield (1974)
 reported  that more than 90% of the radiolabel from an  oral  dose of 2,000
 mgAg   C-labeled DEHP was excreted in the urine. Very little DEHP was
 absorbed  intact. The diester was hydrolyzed by intestinal esterases, and
 the MEHP  is the primary form absorbed (Oishi and Hiraga 1982, White ec
 al. 1980).

     Schulz and Rubin (1973), who administered 250 mg  L4C-DEHP to rats
 in corn oil, found that approximately 61% of the administered dose was
 found in  the organic solvent extracts of urine,  while  12.8% was found in
 Che feces and contents of the large intestine.

     Albro et al. (1982) and Albro (1986) have reported that there is a
 relatively discrete absorption threshold for single oral gavage dosages
 of DEHP in Fischer rats. This may be due to saturation of esterases in
 the gastrointestinal tract.  The animals received 1.8 to 1,000 mg/kg
  C-DEHP  in cottonseed oil.  As the dosage increased, a threshold was
 reached above which there was a steady increase in the amount of
 unhydrolyzed DEHP reaching the liver.  Additional studies utilizing
 dietary administration demonstrated that intact DEHP reached the liver
 at tissue level concentrations exceeding 4,300 ppm. In contrast to the
 results observed in rats,  Albro et al. (1982) did not  detect an
 absorption threshold in either CD-I or B6C3F1 mice administered up to
 1.000 mg/kg DEHP.

     Rhodes et al.  (1986)  reported that,  in the  marmoset,  the excretion
 profile and tissue levels  of radioactivity following oral administration
 of L**C-DEHP demonstrated considerably reduced absorption as compared co
 the rat.  The urinary metabolite pattern in the  marmoset was, in many
 respects,  qualitatively similar to but quantitatively different from
 that in the rat.

     Chadwick et al.  (1982)  demonstrated that 14C-7-DEHP is almost
 completely absorbed when administered in the diet to male F344 rats at
 concentrations of 1,000,  6,000,  and 12,000 ppm for 24 h,  that is,  after
 pretreatment for 0,  6,  or  20 days at these concentrations with unlabeled
 DEHP.

 4.2.1.3  Dermal

     Human.   No data are available on the absorption of DEHP in dermally
 exposed humans.

     Animal.   DEHP appears to be  poorly absorbed when applied to the
 skin of rats.  El Sisi et al.  (1985)  studied the  dermal absorption of
 lftC-DEHP In male Fischer 344 rats over a 5-day  period. The  site of
 application was a shaved area of  skin 1.8 cm in diameter that was
occluded with a perforated cap. The  degree of absorption of DEHP was
 estimated from the  cumulative daily  excretion of 14C-DEHP equivalents in
 the urine  and feces  over 5 days.  Only 5.1% of the administered dose of
 30 mg/kg 14C-DEHP was excreted over  a 5-day period, with 3.0% appearing
 in the urine and 2.1% in the feces.  An additional 1.8% of the
administered dose was found  in the tissues,  with muscle tissue
containing 1.2% of the  administered  dose.

-------
  36   Section 4

      The nature of the absorbed species was not determined. Onlv total
  radioactivity was measured; therefore,  it is unknown whether DEHP was
  metabolized, and if so. to what products. At present,  it is not known
  what happens to DEHP after it is taken up by the skin.  Mammalian skin
  possesses cytochrome P-450 activity,  epoxide hydrolase.  and
  glutathione-s-transferase. and has been shown to metabolize several
  compounds (Bickers et al.  1982.  Albro et al.  1984).  However, the ability
  of the mammalian skin to de-esterify  xenobiotics in vivo has not been
  established, and the existence of nonspecific lipases  and esterases in
  the skin has not been demonstrated (CPSC 1985).

  4.2.2  Distribution

      Absorbed DEHP and its metabolites  are  rapidly distributed to the
 organs and tissues with no apparent accumulation.  The  tissue
 distribution of DEHP and its  metabolites after  oral  dosing appears  to be
 species dependent,  with differences being reported for  the rat,  mouse.
 and monkey.

 4.2.2.1  Inhalation

      Human.   No data were  available on  the  distribution  of DEHP  by
 humans  exposed  via inhalation.

      Animal.  No data were available  on the distribution of DEHP by
 animals exposed via inhalation.

 4.2.2.2  Oral

      Human.  No  data  were  available on  the distribution  of DEHP  in
 orally  exposed humans.

      Animal.  Eriksson  and Darnerud (1985) performed a quantitative
 retention study  of  ^C-labeled DEHP. Groups of mice aged  3,  10   and 20
 days  were orally administered 0.7 mg of  l^C-7-DEHP by gavage  in  a fat
 emulsion. The mice were killed 24 h or  7 days after treatment, and  the
 amount  of radioactivity in the whole brain was measured and  its
 distribution studied. The  retention of DEHP in the brain was  minimal.
 especially in the 10- and  20-day-old mice, where no radioactivity was
 found 7 days after treatment.  The amount of radioactivity  in  the  liver
 was two to three times higher than that  in the brain. Mice exposed  to
 DEHP  showed a significant  decrease in the amount of retained
 radioactivity between 24 h and 7 days  after treatment in all  three age
 categories.

      Pollack et al. (1985b) studied the pharmacokinetics of unlabeled
 DEHP and its biologically active metabolite MEHP in rats following
 single or multiple administration of DEHP by various routes.  DEHP was
 assayed in whole blood by a high performance liquid chromatographic
 (HPLC) procedure. Following a single intraarterial injection, a  large
apparent volume of distribution (5.390 mL/kg) and a high rate of
clearance (21.5 mL/minAg)  were observed for DEHP. In contrast,  the
systemic availability of DEHP was low  following either a single oral
 (13.6%) dose administered as a 40% solution in corn oil, or by
intraperitoneal (5.2%) administration   A marked route dependency in the
formation of MEHP from DEHP was observed. The circulating concentrations

-------
                                                 ToxLcological Daca   37

of MEHP were substantially higher than those of DEHP, with a ratio of
the area under the curve (AUC) for concentration in blood vs time to
that of DEHP of -7 after oral administration. In contrast.
concentrations of the mono-de-esterified metabolite were much lower
relative to the parent diester concentration after intraarterial or
intraperitoneal administration, with an AUC ratio less than 0.4.
Pharmacokinetic modeling indicated that approximately 80% of an oral
dose of DEHP undergoes mono-de-esterification, as compared to only about
1% of the dose following either intraarterial or intraperitoneal
administration. This suggests that the low oral systemic availability of
DEHP may be largely attributed to presystemic hydrolysis of DEHP to MEHP
in the gut, whereas slow and/or incomplete absorption is the likely
cause of the poor bioavailability of DEHP after intraperitoneal
administration. No significant accumulation in the circulating
concentrations of DEHP or derived MEHP were observed following 7 days of
repetitive oral administration of DEHP.  However, multiple
intraperitoneal injections resulted in an apparent decrease in the rate
and/or extent of DEHP absorption from the peritoneal cavity, while no
significant change in the oral absorption of the diester was observed
The striking difference in the MEHP to DEHP AUC ratio between oral and
intraperitoneal routes was still evident after multiple dosing.  These
data suggest that previously reported differences in the biologic
effects of DEHP in rodents following different routes of administration
may be due to route dependency in the mono-de-esterification of the
diester.

     Bratt and Batten (1982) observed clear species and sex differences
in the tissue retention of DEHP.  Rats and marmosets were given 1,960
nig/kg/day 14C-DEHP orally for 14 days.  In the rat,  the females retained
higher concentrations of the L4C-radiolabel in the liver and kidney than
the males.  A similar pattern was observed in the marmoset. The rats of
both sexes retained higher tissue concentrations of ^C-radiolabel than
did the marmosets.

     Similar results were also reported by Rhodes et al. (1986)  in a
comparison of the blood and tissue levels of DEHP and its metabolites in
the rat and marmoset. The animals were administered 2,000 mg/kg/day
l^C-DEHP for 14 days. The test animals were sacrificed 24 h after the
last treatment. The level of l^C-radiolabel in the marmoset tissues was
only 10 to 20% of that in the rat. In both the rat and marmoset, the
liver retained the highest level of 14C-radiolabel.

     Lindgren et al.  (1982) investigated the distribution and tissue
retention of 14C-DEHP (carbonyl-uC or 2-ethylhexyl-l-14C) in pregnant
and nonpregnant mice with whole-body autoradiography. Initially, a high
activity was observed in the brown fat,  liver, gallbladder, intestinal
contents,  kidney, and urinary bladder.  Pretreatment with DEHP,
phenobarbital sodium, or 3-methylcholanthrene caused a relative increase
of the activity in the brown fat, indicating that induced metabolic
conversion of DEHP leads to an increased deposition of radioactivity in
brown fat.  Lindgren et al.  (1982) speculated that pretreatment with DEHP
and other known inducers of mixed-function oxidases caused an increase
in the production of DEHP metabolites with an affinity for brown fat

-------
  38   Section  4

      After administration of DEHP (carbonyl-14C),  but not DEHP (2-
  ethylhexyl-l-14C), marked retention was observed in the skin, cartilage
  and  tendons.  The mechanism responsible for the slow accumulation in
  these connective tissues is not known. In the early embryo, a high
  concentration was observed in the neuroepithelium.  This pronounced
  uptake may be correlated to DEHP-induced exencephaly and spina bifida
  observed  in mice.

  4.2.2.3  Dermal

      Human.  No data are available on the distribution of DEHP'in
  derraally exposed humans.

      Animal.   No data are available  on the distribution of DEHP  in
 dermally exposed animals.

 4.2.2.4  Systemic

      Human.  Sjoberg et al.  (1985a,d)  studied the disposition of  DEHP
 and its primary metabolite  MEHP in newborn infants  subjected to exchange
 transfusions.  During a single  exchange transfusion,  the amounts of
 unlabeled DEHP and MEHP infused ranged from 0.8  to  3.3 and 0.05 to 0.20
 mg/kg,  respectively.  Approximately 30% of the infused  amount of DEHP  was
 withdrawn during the course  of repeated exchange transfusion.
 Immediately after the transfusions,  the plasma levels  of DEHP ranged
 between 5.8 and 19.6 ng/mL,  and subsequently  they declined rapidly. This
 decline,  probably reflecting distribution of  DEHP within the body,  was
 followed by a  slower elimination phase.  The half-life  of this phase was
 approximately  10 h.  The maximal  plasma levels  of MEHP  were about  5
 Mg/mL.  In one  preterra infant,  the  elimination of MEHP  was  slower  than
 its  formation,  whereas  in one  full-term newborn, the formation appeared
 to be rate-limiting  for elimination.

      Pollack et  al.  (1985a)  assessed the  degree of exposure  to DEHP in
 11 patients undergoing  maintenance hemodialysis for the  treatment  of
 renal failure. The amount of DEHP  leached  from the dialyzer  tubing
 during a  4-h dialysis session was  estimated by monitoring  the DEHP  blood
 concentration  gradient  across  the  dialyzer. The plasma concentrations of
 DEHP- and MEHP-derived  metabolites were determined by  gas
 chromatography-chemical ionization spectrometry and gas  chromatography-
 electron  impact  mass  spectrometry, respectively. Circulating
 concentrations of the biologically active  products of  DEHP de-
 esterificaclon,  MEHP  and phthalic acid, were also determined during the
 dialysis  session. On  the average, an estimated 105 mg  of DEHP was
 extracted  from the dialyzer during a single dialysis session,  with  a
 range of  23.8  to 360  mg.  The rate of extraction of DEHP  from the
 dialyzer was correlated with serum lipid content as expressed by the  sum
 of serum cholesterol  and triglyceride concentrations (r  -  +0.65,
 P < 0.05). Time-averaged circulating concentrations of MEHP  during
 dialysis  (1.33 ± 0.58 Mg/mL) were similar  to those of  DEHP (1.91 ±  2.11
Mg/mL).  Blood  concentrations of phthalic acid  (5.22 ±  3.94 /ig/mL) were
higher than those of  the  esters. The length of time patients had been
 receiving regular dialysis treatment was not a determinant of
circulating concentrations of DEHP or MEHP  In contrast, time-averaged
circulating concentrations of phthalic acid correlated strongly with  the

-------
                                                  ToxicoLogical Daca    39

 duration (In years)  of  dialysis  treatment  (r  -  +0.92,  P  < 0 001)  The
 results  indicated substantial exposure  to  DEHP  during  hemodialysis and
 that de-esterification  products  of DEHP are present  in significant
 concentrations  in the systemic circulation.

      Animal.  Sjoberg et al. (1985b) studied  the  disposition of DEHP ard
 four of  its  major metabolites in male rats. The test animals were given
 single intravenous infusions of  a DEHP  emulsion in doses of 5, 50, or
 500  mg/kg  DEHP.  Plasma  concentrations of DEHP and metabolites were
 followed for 24  h after the start of the infusion. The plasma
 concentrations of DEHP- and MEHP-derived metabolites were determined by
 gas  chromatography-chemical ionization  spectrometry and  gas -
 chromatography-electron impact mass spectrometry, respectively. The
 concentrations of DEHP  in plasma were at all  times considerably higher
 than those of MEHP,  and the concentrations of MEHP were much higher than
 those of the  other investigated metabolites.  In animals given 500 mg/kg
 DEHP, the  areas  under the curves (AUCs) for concentration in plasma vs
 time of  the  other investigated metabolites were less than 15% of that of
 MEHP. The  clearance  of  DEHP was much lower in the high-dose group than
 in the lower-dose  groups. This is probably due to saturation of enzymes
 responsible  for  the  hydrolysis of DEHP, product inhibition, or
 saturation of biliary excretion.  Parallel decreases in the plasma
 concentrations of DEHP, MEHP,  and the omega-  and  omega-1 oxidized
 metabolites  indicated that the plasma clearance of DEHP was the rate-
 limiting step in  the disposition of the metabolites. This was partly
 supported by  the  observation that the clearance of MEHP was higher than
 that of  DEHP. Nonlinear increases in the AUCs of  DEHP and MEHP indicated
 saturation in the  formation as well as  the elimination of the
 potentially  toxic  metabolite MEHP.

 4.2.3  Metabolism

     The metabolism  of DEHP involves a complex series of primary and
 secondary pathways.  The proposed metabolic pathway for DEHP is shown in
 Fig.  4.1. The first  step in metabolism  is the lipolytic cleavage of
 DEHP, resulting in the  formation of MEHP and  2-ethylhexanol. Oxidative
 metabolism of MEHP occurs via omega- and omega-1  oxidation of the
 aliphatic side chain. It is believed that this step is followed by a
 dehydrogenase-dependent oxidation to the ketone or carboxylic acid,  with
 subsequent alpha- and beta-oxidation of the acids (Albro et al.
 1983a,b). The metabolism of 2-ethylhexanol appears to be primarily via
 the oxidative carboxylation leading to the formation of 2- and 4-oxy-3-
 carboxyheptane.  The primary urinary metabolites of 2-ethylhexanol are
 2-ethylhexanoic acid and several  keto acid derivatives that appear to be
beta-oxidation products of the primary metabolite.

 4.2.3.1  Inhalation

     Human.  No data were available on the metabolism of DEHP by humans
exposed via inhalation.

     Animal.   No data were available on the metabolism of DEHP by
animals exposed via  inhalation.

-------
                      OEHP
                             ^^
                                             COOCH2CH(C^)3CH3
                                           HYOROUSB (LVMSES)
                    MEHP
                                                                                                                                     l/l
                                                                                                                                     (B
                                                                                                                                     O
                                                                                                                                     rt
    \
OOOH
COOCH,CH|OH^OOCH(
                               OOOCH.CHCOH.Ip.,
                                                \
                                                CH,CH,OM
                                                                    OOOCMjCMICMj^.OM
                                                                                    6-oxB/moN
                                                                                                                CK.CH,
                                                                                                OOOH
                                                                                                 OOOCHjCHCMgCHOHO^COOM
                 O^OM,
                                                                                          COOOMjCHCMjCOOM
               Fig. 4.1.  Proposed melaboliinn for di(2-ethylhexyl)phlhalale in the ml.

-------
                                                   Toxico logical Data   £L
  4.2.3.2  Oral

       Human.   Schraid and Schlatter (1985)  found chat DEHP taken orally  bv
  two volunteers  (30  mg each)  was  excreted  in the urine at 11 and 15% of '
  Cf%cSpr    ter enzy»lc hydrolysis,  the urinary metabolites (derivatives
  of MEHP)  were methylated and identified by  gas liquid chromatography-
  mass spectrometry,  and the quantitative distribution of conjugated  and
  free metabolites was  determined.  Twelve metabolites were  detected   the
  four major ones being free and conjugated forms  of  the  monoester and its
  6-carboxylic  acid.  5-keto. and 5-hydroxy derivatives. The  results of
  this study suggest  that  the  theory that none of  the five major
  metabolites of  DEHP in rats  (Sjoberg et al. 1985a.  1986a)  are major
  metabolites in  man may be incorrect. Schraid and  Schlatter  (1985) found
  that one of the two major metabolites in man involved oxidation of  the
  terminal  (C-6)  methyl  group  to the corresponding acid,  a
  biotransformation process that also occurs in  the rat (Watts 1985)  The
  reason for this is not clear, but may reflect  different dose levels or
  different routes of administration.
  P nc        pLhu8enot ec al- <1985> investigated the in vivo metabolism
 of DEHP and MEHP in rats after multiple dosing. Rats were orally
 administered ^C-DEHP or ^C-MEHP at doses of 50 and 500 mg/kg for 3
 consecutive days. Urine was collected at 24-h intervals, and metabolite
 profiles were determined.  After a single dose of either compound
 urinary metabolite profiles were similar to those previously reported
 However, after multiple administration of both DEHP and MEHP at 500
 mgAg,  increases in omega -/beta -oxidation products [raono(3-carboxy-2-
 ethylpropyl)  phthalate and mono(5-carboxy-2-ethylpentyl) phthalate
 respectively]  and decreases in omega -1- oxidation products [mono(2-
 ethyl-5-oxohexyl) phthalate and mono (2-ethyl- 5 -hydroxyhexyl) -phthalate
 respectively]  were seen.  At the low dose of 50 mg/kg,  little or no
 alteration in urinary metabolite profiles was observed.  At 500 mg/kg
 MEHP, a 4-fold stimulation of cyanide -insensitive palmitoyl-CoA
 oxidation (a  peroxisomal beta-oxidation marker)  was  seen after three
 consecutive daily doses. At the  low dose of 50 mg/kg,  only a 1.8-fold
 increase was  noted.  Similar observations were made by  Lhugenot et  al
 (1985)  with rat hepatocyte  cultures.

     Chadwick  et al  (1982)  treated  male F344 rats with 14C-7-DEHP  in the
 diet at concentrations  of  1.000,  6.000,  and 12,000 ppm for 24 h
 following pretreatment  for  0.  6.  or 20  days with unlabeled DEHP. In the
 animals without prior exposure  to DEHP,  there was a  dose-related,
 nonlinear,  disproportionate  increase  in the fraction of  the  dose
 converted to metabolites I  and V, that  is,  the 3-  and  5-carboxylic acid
 derivatives formed via  the  omega-oxidation  pathway.  The  percentage of
 Increased  urinary excretion of metabolites  I  and V was accompanied by a
 reduction  in the percentage of the  administered  dose excreted in the
 feces.  A  reduction in the fecal excretion of  metabolites  IX  and VI  the
 5-hydroxy  and 5-keto metabolites  formed  via the  omega- 1  oxidation
 pathway, was also reported.

     Prior exposure to DEHP resulted  in  nonlinear  increases  with dose in
 the fraction of the administered dose metabolized via  the  omega- and
beta-oxidation pathways to metabolite I  and excreted in  the  urine.  Prior
exposure to 1,000-ppm DEHP resulted in a quantitative change in the

-------
 42   Seccion 6

 pattern of DEHP metabolism.  After either 6  or 20 days of pretreatment,
 the percentage of the administered dose  excreted in the urine  as
 metabolite I was doubled.  However,  the percentage excreted as  metabolite
 V, the proposed precursor of metabolite  I,  remained unchanged. The
 apparent increase in the conversion of metabolite V to metabolite I may
 have resulted from hepatic enzyme induction.  DEHP is known to  induce
 peroxisome proliferation,  and has been shown  to  induce microsomal
 cytochrome P-450 and 7-ethoxycoumarin 0-deethylase activity in the rat
 (Mangham et al.  1981).  The increased metabolism  via the omega-oxidation
 pathway was offset by a parallel  decrease in  the percentage of
 administered dose converted to the  omega-1  oxidation products,
 metabolites IX and VI,  and excreted in the  urine and feces.  At 6,000 and
 12,000 ppm,  prior exposure to DEHP  for either  6  or 20 days  resulted in a
 major increase in the percentage  of administered dose metabolized via
 the omega-oxidation pathway.  Urinary excretion of metabolite I was
 significantly increased,  accompanied by  a parallel,  dose-related
 decrease in the  urinary excretion of metabolite  V.  This  was  offset by an
 overall decrease in the conversion  of MEHP  to  omega-1 oxidation
 products.

      Agarwal et  al.  (1982)  reported that  the administration of DEHP as a
 single oral  dose of 2.6,  5.2,  or  13.0 mL/kg or a single  intraperitoneal
 dose  of 5.0,  10.0,  or 25.0 mL/kg  significantly increased the
 pentobarbital-induced sleeping time  in rats. Repeated intraperitoneal
 administration of the same doses  for 7 consecutive days  also increased
 the  sleeping time,  but  repeated oral administration decreased  it in a
 dose-dependent manner.  A single exposure  to DEHP by either  route
 significantly inhibited the  activity of aminopyrine  tf-demethylase and
 aniline  hydroxylase  without  any change in the  activities of
 benzo(a)pyrene hydroxylase,  glucose  6-phosphatase,  or NADPH-cytochrome c
 reductase or the concentration of cytochrome P-450 in rat  liver.
 Repeated intraperitoneal administration of  DEHP  had effects  similar to
 those  observed with  a single  exposure. However,  repeated oral doses of
 DEHP  resulted in a  significant increase  in  the activities of aminopyrine
 tf-demethylase, aniline  hydroxylase,  and benzo(a)pyrene hydroxylase,  and
 in  the concentration of cytochrome  P-450. The  activity of glucose 6-
 phosphatase  was  significantly Inhibited in  a dose-dependent manner,  but
 that of  NADPH-cytochrome c reductase remained  unaffected after repeated
 oral and intraperitoneal administration of  DEHP.  Treatment  with DEHP
 failed to affect total  and ascorbate- or NADPH-dependent lipid
 peroxidation as  well as total  and nonprotein sulfhydryl  contents of rat
 liver. Under in  vitro conditions, DEHP had no  effect  on  the  activity of
 aminopyrine  tf-demethylase or  aniline hydroxylase,  whereas mono-(2-
 ethylhexyl)  phthalate and  2-ethylhexanol significantly  inhibited the
 activity of  both enzymes at concentrations  ranging from  2.5  to 15.0 mM.
 the effect of 2-ethylhexanol  being more pronounced.

 4.2.3.3  Dermal

     Human.   No  data were available  on the metabolism of DEHP  in
dermally exposed humans.

     Animal.  No  data were available on the metabolism of DEHP in
dermally exposed animals.

-------
                                                  Toxicological  Data    U1

 4.2.3.4   In  vitro  studies

      Lhugenoc  et al.  (1985) reported  that MEHP  at concentrations  of  50
 and 500 jiM was extensively metabolized  in the rat hepatocyte  cultures.
 Metabolic profiles similar to those seen after  in vivo  administration  of
 MEHP were observed. At the higher concentration of MEHP,  changes  in  the
 relative proportions  of omega- and omega-1 oxidized metabolites were
 seen. Over the 3-day  experimental period, omega-/beta-oxidation produces
 increased in a time-dependent manner  at the expense of  omega-1 oxidation
 products. At a concentration of 500 jiM MEHP, a  12-fold  increase of
 cyanide-insensitive palmitoyl CoA oxidation (a  peroxisomal beta-
 oxidation marker) was observed.  At the low concentration  of MEHP
 (50 /^M), only a 3-fold increase in cyanide-insensitive  palmitoyl-CoA
 oxidation was noted, and little alteration in the metabolite  profile of
 MEHP was observed with time.  Biotransformation  studies  of the
 metabolites  of MEHP confirmed the postulated metabolic  pathways.
 Mono(3-carboxy-2-ethylpropyl)phthalate and mono(2-ethyl-5-
 oxohexyl)phthalate appeared to be end points of metabolism, while
 mono(5-carboxy-2-ethylpentyl)phthalate was converted to mono(3-carboxy-
 2-ethylpropyl)phthalate,  and mono(2-ethyl-5-hydroxyhexyl) phthalate  to
 mono(2-ethyl-5-oxohexyl)phthalate.

     Gollamudi et al.  (1983)  studied the impairment of  DEHP metabolism
 in  young and aged rats.  Liver,  kidney, and lung homogenates from 45- and
 630-day-old  rats were  incubated with 14C-labeled  DEHP.
 Radiochromatograms of  ether extracts of the incubated mixtures from
 young animals showed peaks corresponding to the parent  compound and  the
 hydrolytic product MEHP.  Preparations from old animals  revealed a
 dramatic reduction in  the formation of MEHP in the  liver  homogenates
 This difference may be attributable to differences  in Km  values of the
 enzymes from adult and old rats,  respectively.  Protein  content in the
 three tissues did not  differ  between young and old  animals. Because the
 formation of MEHP is a major  step in the metabolism of  DEHP,  impairment
 of  this conversion could  possibly alter the rate  of its excretion and
 its  toxicologic significance.

 4.2.4  Excretion

     DEHP is  converted principally to polar derivatives of the
 monoesters by oxidative metabolism prior to excretion. Clearance from
 the body is  rapid,  and there  is  only a slight cumulative  potential. The
 major route  of DEHP elimination  from the body is urinary  excretion.

 4.2.4.1  Inhalation

     Human.   No data were available on the  excretion of DEHP by humans
 exposed via  inhalation.

     Animal.   No  data  were available on the excretion of  DEHP by animals
exposed via  inhalation.

4.2.4.2  Oral

     Human.   Schmid and Schlatter (1985)  found that DEHP  taken orally by
 two volunteers  (30  mg  each) was  excreted in the  urine at  11 and 15% of
 the dose.  DEHP  taken by the same  volunteers  for  4 days at 10 mg daily

-------
 44   Section 6

 gave no evidence of accumulation;  15  and  25% of  the  total  dose was
 recovered in the urine.  On the  basis  of this,  the  investigators
 estimated a half-life of 12 h and  concluded that accumulation of DEHP in
 the body is unlikely to  occur.  Unfortunately,  fecal  analysis  that could
 have supported this hypothesis  was not performed.  Generally,  the data
 compare well with those  of Albro et al. (1982) for human leukemia
 patients who received infusions of DEHP-contaminated blood.

      Animal.   Rhodes et  al.  (1986)  studied the comparative
 pharmacokinetics of DEHP in the Wistar rat and the marmoset monkeys
 following oral,  intravenous,  or intraperitoneal  administration.  In both
 the rat and marmoset,  multiple  oral administrations  of  l^C-DEHP at 2,000
 mg/kg did not modify the proportion of dose excreted in the urine or
 feces in either  male or  female  animals.

      Chadwick ec al.  (1983)  administered  100 mg/kg 14C-7-DEHP  by gavage
 to three male Cynomolgus monkeys,  five male F344 rats,  and five groups
 of five B6C3F1 mice.   Urinary and  fecal excretion  of the radiolabel was
 almost complete  after  24 and  48 h,  respectively. The radiolabel in the
 urine and feces  was  resolved  into  as many as 12  components. The
 monoester derivative of  DEHP  was found in the monkey and mouse  urine,
 but was not  detected in  the  rat urine. Metabolite  V,  the 5-carboxylic
 acid omega-oxidation product, was  a major metabolic  product in  rat and
 monkey urine.  Metabolite IX,  the 5-hydroxy omega-1 oxidation product,
 was a major  component  in the  urine  of all three  species. Metabolite VI,
 a  5-keto derivative  formed  from metabolite IX,  was a major component  in
 the urine  of  mice and  rats but  not  monkeys.  Metabolite  I, the  3-
 carboxylic acid  derivative  formed via beta oxidation, was also  a major
 metabolite in the urine  of mice and rats but not monkeys. These  data
 suggest that  beta oxidation  is  a major metabolic pathway in rodents but
 not in nonhuman  primates.

      Chadwick et al. (1982) reported that urinary  excretion of
 radioactivity increased  with  dose   whereas fecal excretion decreased
 with  dose  in  male F344 rats fed L4C-7-DEHP in the  diet  at concentrations
 of  1.000, 6,000, and 12,000 ppm for 24 h following pretreatment  for 0,
 6,  or  20 days with unlabeled  DEHP.   The time  course patterns of
 elimination of DEHP metabolites via the urinary  and  fecal routes  were
 different. Urinary excretion of metabolites  primarily occurred  during
 the  first 24  h after dosing, whereas fecal excretion occurred primarily
 during  the second 24-h phase. The differences  in clearance of
 metabolites are probably due  to the enterohepatic  recycling of
 metabolites secreted via the bile and reabsorbed in  the gastrointestinal
 tract.

     Teirlynck and Belpaire (1985)   studied the  disposition of DEHP and
MEHP  in male Wistar rats. Three hours  after  a  single  oral dose  of 2,800
mg/kg DEHP, plasma concentrations  of 8.8 /ig/mL DEHP  and 63.2 jig/mL MEHP
were observed. The ratio of the area under the  curve  (AUC)  for
concentration in plasma vs time of MEHP to that of DEHP was 16.1.  MEHP
had a half-life of 5.2 h. When  14C-DEHP was  administered, 19.3%  of the
 radioactivity was excreted in the  urine within  the first 72 h. The
remaining radiolabel was excreted in che feces. The  urinary excretion
rate indicated a half-life of 7.9  h  Multiple  dosing with 2.800  rag/kg

-------
                                                  lexicological Daca   ^5

 DEHP for 7 consecutive days produced no accumulation of either DEHP or
 MEHP.

 4.2.4.3  Dermal

      Human.   No data were available  on the  excretion of DEHP in dermal!,
 exposed humans.

      Animal.   No data were available on the excretion of DEHP in
 dermally exposed animals.

 4.2.4.4  Systemic

      Human.   No data were  available  on the  excretion of DEHP in
 systemically  exposed humans.

     Animal.   Metabolites  of  DEHP  found in  urine  from African green
 monkeys after intravenous  administration of ^C-labeled parent compound
 were  isolated and identified  by Albro  et al.  (1981).  Urinary metabolices
 (80%) were excreted  in the form of glucuronide conjugates. This  was
 analogous  to  that reported for the urinary  metabolites of DEHP from
 humans,  but in contrast  to metabolites  found  in rat  urine. Rat urinary
 metabolites of DEHP  were excreted unconjugated and consisted primarily
 of  derivatives more  highly oxidized  than the  major metabolites produced
 by  monkey or  human.  This suggests that  the  African green monkey  may be a
 better  model  for  human metabolism of DEHP than the rat (Peck and Albro
 1982).  Interspecies  differences in the  metabolism of  DEriP are  summarized
 in  Fig.  4.2.

 4.2.5   Discussion

     Animal experiments have  shown that  DEHP  is readily  absorbed after
 ingestion. In  the one  study available  (El Sisi et al.  1985),  it  was
 shown that substantially less DEHP is absorbed through the skin, and che
 nature  of the  absorbed species is unknown.

     Oral exposure to  less  than very high dose levels  of DEHP  does  noc
 result  in tissue  exposure  to  the intact  diester, which is rapidly and
 efficiently hydrolyzed in  the gastrointestinal tract by  microflora and
 hydrolytic enzymes.

     The primary metabolites,  MEHP and  2-ethylhexanol, are rapidly
 oxidized to a variety of more polar products. The biological activity of
 the metabolic products has not been investigated extensively.  There are
major interspecies differences in the metabolism of DEHP.

     In rodents (rats and mice),  the excreted metabolites of DEHP
primarily consist of diacids and ketoacids,  which are  terminal oxidation
products. However, in both humans and the African green monkey,  the
major excretion metabolites are MEHP and minimally oxidized hydroxyacid
products.

     In addition to interspecies  differences, there are major
qualitative differences in metabolism and major quantitative differences
in excretion rates between young  and old animals of the  same species

-------
                          — OR
      DEHP
   HYDROLYSIS
RODENTS
  AND
PRMATES
      MEHP
GLUCURONIDATION
      AND
  CO andcj-1
  OXDATIONS
                          — OH
        CH2CH3

        I
R - -CH2CH2(CH2);jCH3

R'-OXDATION PRODUCTS OF R
                                      RATS
                       RODENTS (MICE)
                            AND
                          PRIMATES
                                          — OH
                                                               — OR
                                         — O-GLUCURONIDE
                                                                            10
                                                                            (6
                                                                            n
                                                                            n
                                                                            F-
                                                                            o
                                                                            3
                Fig. 4.2.  Species-specific metabolism of dK2-ethylhexyl)phlluilate.

-------
                                                  ToxicologicaL Daca   ^.7

 4.3  TOXICITY

 4.3.1  Lethality and Decreased Longevity

 4.3.1.1  Inhalation

      Human.   Thiess et al.  (1978a) conducted a morbidity study and a
 mortality study (1978b) on 101 workers who had been employed for periods
 ranging from 4 months to 35 years in a DEHP production facility  Currenc
 exposures in work areas ranged from 0.0006 to 0.01 ppm.  but may have
 been higher  prior to 1966 when a process change was made.  Blood,  liver,
 and urinary  conditions were examined,  and for the six worker-s with more
 than 20 years of service,  a neurologic examination was administered
 Analyses of  absenteeism,  of accidents,  and of a reproduction
 questionnaire were conducted.  In-house control groups (with possible
 exposure to  styrene and dimethyl carbamoyl chloride)  were  compared with
 the DEHP workers.  No findings  of morbidity attributable  to DEHP exposure
 were reported.  The lack of  good exposure data and of  a control  group not
 exposed to industrial chemicals limits the utility of this morbidity
 study.

      In the  mortality study (Thiess  1978b),  which involved 221  workers
 with average time  in service of 11.5 years,  no increase  in mortality
 with increase in exposure duration was  seen.  A single case of bladder
 papilloma resulted in a higher-than-expected rate for that phenomenon.
 but this  was not considered sufficient.

      Thiess  et  al.  (1979) also  performed chromosomal  analysis on  blood
 lymphocytes  from a subset of this  same  study population. Lymphocytes
 were  cultured from 10 exposed production workers  according to a modified
 method  of Moorhead et al. (1960).  The workers'  duration  of exposure
 ranged  from  10  to  34  years  (mean - 22.1  years). Lymphocytes  from  20
 age-matched  workers  served  as controls.  It was  not  stated  whether these
 controls  were also  exposed  to styrene and DMCC, as  mentioned previously
 One hundred  metaphases  were scored for  abnormalities  on  lymphocytes  from
 each  worker.   The specific structural abnormalities  were  not  defined,  buc
 were  categorized with and without  gaps.  Neither category appeared to be
 different from  the  control, although statistical  analyses  were  not
 stated.

      None of  the above  three studies by  Thiess  et al.  can  be  used to
 establish a NOAEL.

     Animal.   Acceptable lethality data  were not  available  for  animals
 exposed to DEHP. In an  acute rat study.  1 h exposure  to  23,670  mg/m3
 (1,457 ppm) DEHP did  not result in any deaths  (WARF Institute 1976).  In
 a study by Pegg (1979). no deaths were observed in  rats  exposed to 600
 mg/mj (37 ppm) DEHP for 6 h.

 4.3.1.2  Oral

     Human.   No data were available on the effects of  ingestion exposure
 to DEHP on lethality or decreased  longevity in humans.

     Animal.   A number of animal studies, which are summarized  in Table
4.1. have been performed to investigate  the acute toxicity of DEHP   The
LD50 for DEHP ranges from 26.000 to 49.000 mg/kg.  which  indicates thac

-------
Section
                     Table 4.1. Acute lethality of diiZ-ethylhexvliphthalaie
Route of
administration
Inhalation
(1 h)°
Inhalation
(6h)°
Oral




•


Inirapentoneal



Intravenous
Intravenous
(sonicated in
rat serum)
Species
Rat
Rai
Rat
Rat
(Wisiar. male)
Rat
(Wistar. male)
Mouse
Mouse
Mouse
Guinea pig
Rabbit
Rat
Rat
(Wistar. male)
Mouse
Mouse
(ICR. male)
Mouse
Rat
LD50
(mg/kg)


26.000
>34000
30.600
49.000
26.000
33.300
26.300
33.900
49.000
30.600
4.200
38.000
1.060
2.080
LC50
(mg/m ) References
>23.670 WARF Institute 1976
(1457 ppm)
>600 Pegg 1979
(37 ppm)
Patty 1967
Hodge 1943
Shaffer et al I94S
Yamada 1974
Patty 1967
Krauskopf 1973
Krauskopf 1973
Shaffer et al I94S
Singh etal 1972
Shaffer ei al I94S
Galley etal 1966
Lawrence et al 1973
Petersen et al 1974
Petersen et al 1974
          'Duration of exposure
          Source. Adapted from NTP 1982

-------
                                                   ToxicologicaL Daca   69

  DEHP has  a  low order of acute oral  coxicicy.  Studies  using  the
  intravenous and  intraperitoneal routes of administration, which are also
  summarized  in Table 4.1. provide additional evidence  of  the low order of
  the acute toxicity of DEHP.

      The NTP (1982) evaluated the carcinogenicity of  DEHP in a standard
  bioassay using F344 rats and B6C3F1 mice of both sexes.  In the rats, no
  significant trends in mortality were observed in the  treated animals
  when compared to the controls. In the low-dose female mice, a
  significantly shortened survival was observed compared with the
  controls. However, no positive trend was observed since  there was a
  somewhat longer survival in the high-dose females.

  4.3.1.3  Dermal

      Human.   No data were available on the effects of dermal exposure  co
 DEHP on lethality or decreased longevity in humans except for those
 reported in Sect. 4.3.1.1.

      Animal.  There were no available data on the potential lethality  or
 decreased longevity associated with dermal exposure of animals to DEHP

 4.3.1.4  Discussion

      The available data  suggest  that DEHP has  a  low order of acute oral
 toxicity.  In oral studies  in rodents,  the LDso ranges  from 26,000 to
 49.000  mg/kg.  Data from  a  2-year oral carcinogenicity  bioassay indicate
 that  DEHP does  not induce  dose-related increases  in mortality  in  male
 rats  or mice at doses of 322 or 674  mg/kg/day  and 672  or  1,325
 mg/kg/day, respectively, or in  female rats or  mice  at  doses  of 394 or
 774 mg/kg/day and 799 or  1,821 mg/kg/day.  respectively.

 4.3.2   Systemic/Target Organ Toxicity

     The primary  target  organs of DEHP have  been  shown to be  the  liver
 and testes.

 4.3.2.1  Hepacic  toxicity

     Certain phthalate esters, including  DEHP. and hypolipidemic  agents
 are known  to induce morphological and  biochemical changes in the  liver
 of rodents. Such changes have been associated  with  increased incidences
 of hepatocellular tumors in  these species. While  there is evidence that
 some hypolipidemic agents do not induce these  effects  in  either subhuman
 primates or man (Cohen and Grasso 1981. Lake and Gray  1985). Jacobson ec
 al. (1977) and Canning et al. (1984) contradict this.

     Inhalation, human.   No data were available on the hepatic toxicity
 of DEHP  in humans exposed via inhalation.

     Inhalation, animal.   No data were available on  the hepatic toxicity
 of DEHP  in animals exposed via inhalation.

     Oral, human.   No data were available on the hepatic  toxicity  of
DEHP in orally exposed humans.

     Oral, animal.  Gollamudi et al  (1985) reported that DEHP inhibited
UDP-glucuronyl-transferase activity of rat liver in vitro and  in vivo

-------
 50   Section  4

 Diechyl phthalate  and  dimethoxyethyl phthalate also  Inhibited this
 enzyme  in vitro. Conversely, DEHP did not inhibit the activity of the
 cytosolic enzyme N-acetyltransferase or alter the levels of rat liver
 microsomal  cycochrorae  P-450  in vitro. It is suggested that DEHP may
 alter the composition  of microsomal phospholipids.

     Rhodes et al.  (1986)  reported that oral and  intraperitoneal
 administration of  DEHP to  the marmoset monkey at doses up to 5 mmole
 (1950 mg) DEHP/kg/day  for  14 days did not induce morphological or
 biochemical changes  in the liver comparable with  those obtained in rats
 administered  the same  amount of DEHP.

     In a study of the hepatic effects of DEHP (CMA  1984), groups of
 five male and five female  rats were treated with 0.  0.1, 0.6, 1.2, or
 2.5% DEHP in  the diet  for  3 weeks. In rats fed 2.5%  DEHP, body weight
 and food intake were significantly reduced.  Statistically significant
 increases in  liver weight were observed at dose levels of 0.6, 1.2,  and
 2.5%. There was a  dose-related increase in cyanide-insensitive palmitoyl
 CoA oxidation (pCoA) at dose levels of 0.6% and above. An increase in
 lauric acid 12-hydroxylation was seen at doses of 0.1% and above in the
 males and at  the 1.2 and 2.5% dose levels in the  females. A less-marked
 increase in 11-hydroxylation was also observed. Electron microscopy
 revealed a dose-related increase in peroxisomes at levels of 0.1% and
 above in males and at  0.6% and above in the females.

     The Chemical  Manufacturers Association (CMA) (1985a,b,c,d;
 1986a,b,c,d)  also  evaluated the hepatic effects of eight other phthalate
 esters in rats. Groups of  five male and five female  rats fed 1.2% DEHP
 in the diet for 3  weeks served as positive controls  in each study.
Analysis of the DEHP-positive control values between studies indicates
 that male rats treated with 1.2% DEHP in the diet gained less weight
 than concurrent untreated control male rats. However, the body weight
values observed in the DEHP-treated rats were generally not more than
 10% less than untreated control values. There was substantial
variability (2.38-fold) between the values obtained  for pCoA, a primary
marker for peroxisome  proliferation. Values obtained for 11- and 12-
 lauric acid hydroxylase were also variable (2.28- and 2.33-fold,
 respectively).

     Parmar et al.  (1985)  investigated the effects of DEHP on the
detoxification mechanisms of xenobiotics in neonatal rat pups of mothers
who received  2,000 mg/kg phthalate daily from day 1  of birth to day 21.
The nursing rat pups of mothers exposed to DEHP showed decreases in body
weight gain and activities of aniline hydroxylase, ethylmorphine N-
demethylase,  and arylhydrocarbon hydroxylase, and decreased levels of
cytochrome P-450 at 21 days of age. Significant quantities of DEHP were
also detected In the liver of pups In this Interval. These results
suggest that  the livers of developing animals can be affected by the
lactational transfer of DEHP.

     F.  E.  Mitchell et al. (1985) fed groups of male and female Wistar
albino rats levels of  50, 200, or 1.000 mg/kg/day DEHP in the diet. Four
rats from each experimental group and six control rats of the same sex
were killed 3, 7,  14,  and 28 days and 9 months after the start of
treatment.  At all  time points the major abdominal organs were removed
and subjected to histological examination  A more extensive necropsy was

-------
                                                  TOXLCOlogical  Daca    5L

 performed on chose  rats killed  after  9  months  of treatment.  Two early,
 transient alterations were observed.  There  were  morphologic  changes  in
 the bile  canaliculi  of male rats treated with  1,000  mg/kg/day DEHP.  In
 addition,  the  liver  cells exhibited a burst of mitosis  immediately afcer
 the start of administration of  the compound. The time course of this
 mitotic burst  varied; the increase in mitosis  was  greatest at 3 days  in
 rats treated with 1,000 mg/kg/day DEHP, and was  smaller, but more
 prolonged,  in  rats treated with 50 or 200 mg/kg/day. Other changes,
 including  a  midzonal to periportal accumulation  of fat,  induction of
 peroxisomal  enzymes, and induction of the P-450  isoenzyme, also
 developed  rapidly and were observed throughout the study. The maximal
 change was usually attained within 7  days of the start of treatment.
 More  slowly  developing changes were hypertrophy  of the hepatocytes,
 centrilobular  loss of glycogen,  and a fall  in glucose-6-phosphatase
 activity. Maximal changes were not attained until  28 days after
 treatment began. These effects were clearly observed in  rats treated
 with  200 or  1,000 mg/kg/day DEHP,  but were  only marginally altered in
 rats  treated with 50 mg/kg/day.  Finally, accumulation of lipid-loaded
 lysosomes assessed by light and electron microscopy and by assay of
 beta-galactosidase activity was  only apparent in rats treated with DEHP
 for 9 months with 200 or 1.000 mg/kg/day DEHP.  Changes in female rats
 were  qualitatively similar to those observed in male rats. The
 alterations were, however,  less  pronounced  than  in male  rats treated
 with  an equal dose of DEHP,  and the degree  of liver enlargement was much
 less  because, although the initial hyperplasia was clearly apparent,
 there was a much smaller degree  of hypertrophy.

      Melnick and Schiller (1985) exposed isolated rat liver mitochondria
 to mono- and di-n-butyl phthalate  (MBP and  DBP) and MEHP and DEHP. These
 were  examined for effects on mitochondrial  energy-dependent processes,
 including oxidative phosphorylation and active K+ uptake. Additional
 studies on the effects of these  phthalate esters on succinate oxidation
 and on mitochondrial membrane integrity were also performed.  DEHP had no
 apparent effect on any of these  processes except for slight impairment
 of ATP-dependent £+-valinomycin-induced swelling. The MEHP was  found co
 act as a noncompetitive inhibitor  of succinate dehydrogenase activity
 At concentrations which uncoupled  energy linked reactions, MEHP produced
 slight energy-dependent swelling and the release of soluble proteins
 from  isolated mitochondria,  probably due to an increase in membrane
 permeability to H+ and other small  ions.

      In a study by Carpenter et  al.  (1953),  groups of 23 to 24  guinea
 pigs of each sex were fed diets  containing DEHP at dose levels  of 19 or
 64 mg/kg/day for a period of 1 year.  No treatment-related effects were
 observed on mortality,  body  weight,  kidney weight, or gross pathology
 and histopathology of kidney,  liver,  lung,  spleen, or testes. A
 statistically significant  increase  in relative  liver weight was observed
 in both groups  of treated females.

     Carpenter  et al. (1953)  also  performed a study with groups of male
 and female Sherman rats.  However,  this study was inadequate because of
high mortality  in the control  group,  thus  limiting its utility.

-------
 52    Section  4

      Doscal et  al.  (1987)  investigated the relative sensitivity of
 suckling rats,  as compared to adults, to the effects of DEHP. Five daily
 oral  doses of 0, 10,  100,  1,000, or 2,000 mg/kg DEHP were administered
 to  male  Sprague-Dawley  rats beginning at 6, 14, 16, 21, 42, and 86 days
 of  age.  Twenty-four hours  after the last dose, rats were sacrificed, and
 plasma cholesterol and  triglyceride levels and the activities of the
 hepatic  peroxisomal enzymes, palmitoyl CoA oxidase and carnitine
 acetyltransferase, were determined.

      Suckling rats (1 to 3 weeks of age) suffered severe growth
 retardation at  doses of 1,000 mg/kg and death at 2,000 mg/kg, while
 older rats only showed decreased weight gain at 2,000 mg/kg. Of '
 particular interest was the lethality at doses of 1,000 mg/kg at 14 days
 of  age,  but not at 16 days or at other ages.  Increases in relative liver
 weight and hepatic peroxisomal enzyme activities were similar in all age
 groups except the 14-day-old group, in which the increases were greater.
 Relative  kidney weight was increased in 21-,  42-,  and 86-day-old rats,
 but not  in younger rats, at the highest doses. Hypolipidemia was
 observed  only in 21-, 42-, and 86-day-old rats at doses of 1,000 and
 2,000 mg/kg,  whereas elevated plasma cholesterol levels were observed in
 6- and 14-day-old rats at  the 1,000-rag/kg dose, possibly due to the
 dietary differences between suckling and weaned rats.  The results
 suggest  that  neonatal and suckling rats are more sensitive to the lethal
 and growth retardation effects of DEHP than are adult rats, but the
 hepatic peroxisome proliferation was similar at all ages with the
 exception of  a  greater increase at 14 days of age.

      Dermal,  human.   No data were available on the hepatic toxicity of
 DEHP  in dermally exposed humans.

      Dermal,  animal.   No data were available on the hepatic toxicity of
 DEHP  in dermally exposed animals.

      Systemic, human.  Canning et al.  (1984)  reported that liver
biopsies taken  from dialysis patients showed peroxisome proliferation.

      Systemic, animal.  Walseth et al.  (1982) administered DEHP via
 intraperitoneal injection (3.8 mM/kg)  to Sprague-Dawley rats for 5 days.
DEHP  increased significantly the liver concentration of cytochrome
P-450 but decreased the lung concentration of cytochrome bS and NADPH-
cytochrome-c-reductase activity. The direction of benzo(a)pyrene
metabolism was changed,  and the formation of 2- and 3-hexanol
metabolites was increased in liver microsomes after treatment. The
cytochrome P-450 enzyme system in the lung was 10-fold more effective
than  that in  the liver as far as metabolism of n-hexane was concerned.
Only minor effects  were observed in serum enzyme activities, but a
significant decrease in the serum level of albumin was observed after
treatment with DEHP.  No relationship was found between the carbon chain
length of the investigated chemicals and effects on microsomal enzymatic
activities.

     Discussion.  Exposure to DEHP has resulted in adverse hepatic
effects  in laboratory test animals  DEHP at high concentrations can
cause functional hepatic damage, as reflected by morphological changes.
and alterations in  the activity of energy-linked enzymes and metabolism
of llpids and carbohydrates.  Several studies  have demonstrated that DEHP

-------
                                                  TaxLCOLogical Data   53

 is a peroxisome proliferator.  These observations are significant because
 of an association between peroxisome proliferation and both liver
 hyperplasia and carcinogenesis in laboratory animals  Administration of
 DEHP orally ac sufficient doses has been shown to induce proliferation
 of peroxisomes, increase mitochondria and endoplasmic reticulum
 membranes,  induce cytochrome P-450, and induce hepatomegaly associated
 with hepatic hyperplasia.  Studies performed to date have not established
 any single  metabolite  or metabolic pathway as essential for peroxisome
 proliferation.  In addition,  there have been no long-term studies that
 allow a precise estimate of  the relationship between DEHP dose and
 peroxisome  proliferation.

 4.3.2.2  Testicular  toxicity

      The testicular  effects  produced by DEHP are discussed in the
 section on  reproductive  toxicity.

 4.3.3  Developmental Toxicity

      The available data  indicate  that  DEHP  is  developmentally toxic  in
 rats  at relatively high  doses.  In mice,  DEHP is  developmentally toxic ac
 levels  where  no effects  were observed  in the treated dams.

 4.3.3.1  Inhalation

      Human.   No data were available  on the  developmental  toxicity of
 DEHP  in humans  exposed via inhalation.

      Animal.  No data were available on the  developmental  toxicity of
 DEHP  in animals  exposed  via  inhalation.

 4.3.3.2   Oral

      Human.  No data are available on  the developmental  toxicity  of  DEHP
 in orally exposed humans.

      Animal.  Nakamura et al.  (1979) investigated the  teratogenicity of
 DEHP  in  ddy-Slc x CBA mice. The animals  received a  single  oral
 administration of 0.05, 0.1,  1.0,  2.5,  5.0,  or 10.0  mL/kg  DEHP  by gavage
 on day  7 of gestation.  A dose of 0.05 mL/kg  DEHP resulted  in  a  decrease
 in body weight of live fetuses. However, there were  no  fetal  deaths  or
 soft-tissue or skeletal abnormalities. At doses of 0.1 mL/kg  and  above,
 DEHP  decreased fetal body weight, and  the fetuses were deformed or dead

      Wolkowski-Tyl et al. (1984a)  fed  timed pregnant Fischer  344  rats
 diets which contained 0,  5,000. 10.000.  15.000, or 20,000  ppm DEHP on
 days  0 to 20 of gestation. Based on food consumption data,  this was
 equivalent to doses of approximately 0,  357. 666, 856, or  1.055
 nig/kg/day. There was no maternal mortality  in any of the dosed  animals
 There was a significant dose-response trend  toward maternal body  weight
 loss, decreased gravid uterine weight, and  increased absolute and
 relative liver weights. Clinical signs of toxicity,  including
 piloerection, rough coat, and reduced food  intake, were observed  in  a
 dose-related manner.  There were no dose-related differences observed in
 the number of corpora lutea or implantation sites per dam  or  percentage
of preimplantation losses.  The number and percentage of resorptions,
nonlive fetuses, and affected (nonlive and malformed) fetuses were

-------
 54    Section  4

 Increased In  a  dose-related manner, with a statistically significant
 increase  in all three  parameters  in the high-dose group when compared
 with  the  controls.  The number of  live fetuses per litter exhibited a
 dose-related  decrease,  with the high-dose group significantly lower than
 the controls. Among the live litters, there was a dose-related decrease
 in the body weights of both sexes at all doses tested. There was a
 significant upward  dose-related trend in the percentage of malformed
 fetuses per litter.  However, there were no significant differences in
 pairwise  comparisons in the number of malformed fetuses per litter. The
 results of this study  demonstrate that DEHP is maternally and
 developmentally toxic.  DEHP has been shown to cross the placenta in rats
 (Singh et al. 1975), and the fetotoxicity observed in this study may
 have  been the result of direct contact to the parent compound and/or its
 metabolites with embryonic or fetal tissues.

      Wolkowsky-Tyl  et  al. (1984b) exposed timed pregnant CD-I mice on
 days  0 to 17 of gestation to diets containing 0, 250, 500, 1,000. or
 1,500 ppm DEHP.  Based  on food consumption data, this was equivalent to
 approximately 0, 44, 91,  191, or 292 mg/kg/day. There was no maternal
 mortality in any dose  group. There was a significant dose-response trend
 toward reduced  maternal body weight on-days 12, 16, and 17 of gestation
 in the groups receiving 1.000 or 1,500 ppm DEHP. Gravid uterine weight
 exhibited a dose-related decrease in the 1,000- and 1,500-ppm dose
 groups. Maternal liver  weight exhibited a dose-related increase in the
 two highest dose groups when compared to the controls. Clinical signs of
 toxicity,  which included piloerection, lethargy, and rough coat, were
 seen  in all of  the  DEHP groups in a dose-related manner.

      There  were  no  dose-related differences observed in the number of
 corpora lutea or implantation sites per dam or in the percentage of
 preimplantation losses.  The number and percentage of resorptions [dead.
 nonlive (dead plus  resorbed), and affected (nonlive plus malformed)] per
 litter were increased  in a dose-dependent manner. These parameters were
 significantly higher than the controls for the 1,000- and 1,500-ppm dose
 groups. In  the  1,000- and 1,500-ppm dose groups, there was a
 statistically significant decrease in the number of live fetuses and che
 number of males  and  females per litter.  There was a dose-related
 decrease  in the  body weights of the male and female fetuses. For the
 1,000- and  1,500-ppm dose groups,  the differences were statistically
 significant when compared to the controls.  There were significant dose-
 related increases at the 500-,  1.000-. and 1,500-ppm doses in the number
 and percentage  of fetuses malformed per litter. The major malformations
 included  external, visceral, and skeletal defects. The NOAEL for DEHP in
 this  study  was  250 ppm,  where there was no significant maternal or
 developmental toxicity.

      Tomita et  al.  (1986) have shown that MEHP, one of the major
metabolites of  DEHP, exhibits developmentally toxic effects. Mono(2-
 ethylhexyl)phthalate was given to mice as a single oral dose of 0.1,
0.5.   or 1 raL/kg  MEHP on day 7,  8.  or 9 of gestation. High-dose mice
 treated on  days  7 or 8  exhibited only 18.5 and 31.8% fetal survival at
parturition, respectively. In the latter group. 100% of the live fetuses
were  malformed.

-------
                                                  ToxicoLogLcaL Data   55

      Shioca et al. (1980) demonstrated that DEHP is developmentally
 toxic in ICR-JCL mice. Pregnant animals were given diets containing 500
 1.000. 2.000, 4.000.  or 10.000 ppra DEHP throughout gestation. The mean
 daily intake calculated from food consumption was 70, 190, 400. 830, and
 2.200 rag/kg, respectively.  Treatment with 2,000, 4,000,  and 10,000 ppm
 DEHP resulted in decreased maternal weight gain and increased resorpcion
 rate. At the 4,000- and 10,000-ppm levels, all of the implants died. A
 dose-related pattern of intrauterine growth retardation and delayed
 ossification was observed.  The malformation rate of fetuses at the
 2,000- and 10.000-ppm dose  levels was increased significantly compared
 to controls. The most predominant malformations were neural tube defects
 (exencephaly and spina bifIda).  which suggests that DEHP may interfere
 with the closure of neuropores in the developing embryo.

      Shiota and Mima  (1985)  reported that  DEHP was  developmentally toxic
 in ICR mice when administered orally,  but  not  when  administered by
 intraperitoneal injection.  Groups of pregnant  mice  were  administered
 250,  500,  1,000.  or 2.000 mg/kg  orally or  500,  1,000.  2,000,  4,000,  or
 8,000 mg/kg by  injection on days 7,  8.  or  9  of gestation.  In the groups
 treated  orally,  resorptions  and  malformations  were  significantly
 increased  at the  two  highest dose levels.  Anterior  neural  tube defects
 (anencephaly and  exencephaly)  were  the  most  predominant  abnormalities
 Fetal weights were  also significantly  reduced.  In the  animals treated  by
 intraperitoneal  injection,  DEHP  was  abortifacient and  lethal  to the
 pregnant females  at the higher dose  levels.  However,  no  teratogenic
 effects were observed. Differences  in  the  metabolism,  disposition, or
 excretion  resulting from the  different  routes  of administration may  be
 responsible  for the differences  observed.

      Yagi et al.  (1980) evaluated the  teratogenic potential of DEHP  in
 ddY-Slc mice. Groups  of mice  received a single  oral  administration of  L
 to 30 mL/kg  DEHP  on days 6.  7. 8. 9. or 10 of  gestation. Gross  and
 skeletal abnormalities occurred  in  live fetuses  from  the 2.5-  and  7  5-
 mLAg groups treated  on days  7 or 8 of gestation. The  gross
 abnormalities included exencephaly and club  foot. Skeletal abnormalities
 occurred in  the skull, cervicle,  and thoracic bones. Similar  effects
 were  observed in animals treated with 0.5 or 1.0 mL/kg DEHP on  day 8 of
 gestation.  •

 4.3.3.3  Dermal

     Human.  No data were available on the developmental toxicity of
 DEHP  in dermally exposed humans.

     Animal.  No data were available on the developmental toxicity of
 DEHP  in dermally exposed animals.

 4.3.3.4  Discussion

     The potential developmentally toxic effects of DEHP have been
evaluated in several species. DEHP is developmentally  toxic in both the
 rat and mouse.  In addition,  the metabolite MEHP is developmentalIv
 toxic.                                                           y

-------
 56    Section  4

 4.3.4  Reproductive  Toxicity

      DEHP  has been shown to be a reproductive toxicant in CD-I mice and
 Fischer 344 rats. In other studies. DEHP has been shown to damage the
 cestes.  The testicular  effects produced by DEHP in the rat are
 characterized by a decrease in the relative organ weight and
 histological  changes in the seminiferous tubules (Gangolli 1982).
 Mono(2-ethylhexyl)phthalate, the corresponding raonoester of DEHP formed
 in vivo as a  result  of  the action of nonspecific esterases in the
 intestinal mucosa and other tissues, is equally effective in inducing
 testicular damage. DEHP-induced testicular injury is accompanied by a
 decrease in the zinc content in the gonads and in increased urinary
 excretion of  this element. Exposure of preparations of rat seminiferous
 tubule  cells  in culture to MEHP results in a dose-related detachment of
 germinal cells from  Sertoli cells in a manner similar to the effect seen
 in the  intact animal. The testicular effects observed in the rat and
 mouse have not been  seen in the hamster, ferret, or marmoset (Gray et
 al. 1982. Lake et al.   1976, Rhodes et al.  1986, Jaeckh et al.  1984).

 4.3.4.1  Inhalation

     Human.   No data were available on the reproductive effects of DEHP
 in humans exposed via inhalation.

     Animal.  No data were available on the reproductive effects of DEHP
 in animals exposed via  inhalation.

 4.3.4.2  Oral

     Human.   No data  were available on the reproductive effects of DEHP
 in orally exposed humans.

     Animal.  In a study performed by Reel et al.  (1982),  DEHP was a
 reproductive  toxicant in male and female CD-I mice,  as evidenced by a
 decreased fertility  index, decreased number of litters, decreased number
 of live pups,  and a  lower proportion of live pups per litter.  Groups of
 mice were given 0,  100,  1,000,  or 3,000 ppm DEHP in the diet during a
 7-day premating period  and a 98-day cohabitation period.  Fertility was
 completely suppressed in the high-dose group and significantly reduced
 in the  intermediate-dose group.  In animals receiving 1,000 ppm, the
breeding pairs produced  fewer litters and had fewer male and female live
 pups per litter than  did the controls.  In a crossover mating trial, the
 proportion of detected mating did not differ significantly among
 treatment groups.  However, fertility was significantly reduced
 (P < 0.01)  in both the high-dose male/control female crossover group and
 the control male/high-dose female group. Histological examination of che
high-dose males showed  that the percentage of motile sperm and the sperm
concentration were significantly (P < 0.01) reduced compared with the
controls. In addition,  testicular and epididymal weights were reduced.
and extensive destruction of the seminiferous tubules was observed. In
 the high-dose females,  reproductive tract weights (ovaries, oviducts,
uterus,  and vagina)  were significantly (P < 0.05) decreased when
compared with controls.

     Price  et al.  (1986) evaluated the reproductive effects of DEHP
administered  to time-mated Fischer 344 rats on gestational days 0 to 20

-------
                                                  ToxicoLogical Data   57

 The treatment groups received 0. 2,500. 5.000, or 10.000 ppm DEHP in the
 diet, with resultant average doses of 0, 162.5, 310.4, or 573.4
 mg/kg/day DEHP. During treatment, maternal food consumption and weight
 gain were reduced in a dose-related manner. In the mid- and high-dose
 groups, food consumption was significantly lower than controls. However,
 maternal body weight gain was significantly reduced only in the high-
 dose animals.

      No treatment-related effects were observed in the number of
 implantation sites per dam,  in the percentage of fertile matings,  the
 percentage of live litters,  or the percentage of viable litters.
 Postimplantation mortality was increased in the mid- and high-dose
 groups, but this effect was  statistically significant only at the  middle
 dose.  On postnatal day 1.  average litter size and average pup body
 weight per litter were decreased in a dose-related manner.  In the  high-
 dose group,  the effect on pup body weight was statistically significant.

      During the period between postnatal day 4 through sacrifice,  the
 postnatal growth,  viability,  age of acquisition for developmental
 landmarks,  and levels  of spontaneous locomotor activity of the  Fl
 litters from DEHP-treated dams were comparable to  controls.  Treatment
 with DEHP did not  produce  any adverse effects in the reproductive
 performance  of the Fl  generation or upon the growth and viability  of  the
 F2a litters.

     Agarwal  et al.  (1986)  investigated the sensitivity of sexually
 mature  rats  to the toxic  responses  induced  by the  dietary administration
 of  DEHP.  Adult male  F344  rats  were  exposed  to 0.  320,  1.250,  5.000, or
 20,000  ppm DEHP for  60  days.  Administration of DEHP  resulted in a  dose-
 dependent reduction  in  total  body weight,  testis,  epididymis, and
 prostate weights at  the  two highest dose  levels.  In  the testis,
 degenerative  changes, decreased  testicular  zinc content,  reduced
 epididymal sperm density and motility,  and  increased occurrence of
 abnormal sperm were  observed at  20,000  ppra  DEHP. At  doses of 5,000 and
 20,000  ppm, there  was a  trend  towards  reduced testosterone  and  increased
 luteinizing hormone  and  follicle-stimulating  hormone.

     On day 61  of  the study,  the  treated males  were  returned to a  normal
 diet and mated with  untreated  females.  The  incidence  of pregnancy, mean
 litter weight  on day 1, frequency of  stillbirths and neonatal deaths,
 and mean litter growth up to 7 days of  age  were unaffected by DEHP
 treatment. However,  litter size was significantly reduced in the
 20,000-ppm group.  Cessation of treatment resulted in  partial  to complete
 recovery from  toxicity. The magnitude of recovery was  variable, with
 that of Che gonads being slower  than other  systems. Agarwal  et  al.
 (1986) concluded that these data suggest a  lack of reproductive
dysfunction in the treated male after exposure  to DEHP at levels below
 20,000 ppm, which produced measurable testicular degeneration and
afflicted epididymal sperm morphology.

     Parmar et al. (1986) reported  that oral administration  of DEHP to
adult rats at dose levels of 250. 500.  1.000, and 2,000 mg/kg for  15
days caused a significant dose-dependent decrease in  the  sperm count  of
the epididymal spermatozoa. The activity of gamma-glutamyl
transpeptidase and lactate dehydrogenase was significantly  increased  in
the animals of the treated groups  An increase  in the  activity of  beta-

-------
58   Seccion 4

glucuronidase and decrease in the activity of acid phosphatase were also
observed at the highest dose of DEHP. The activity of sorbitol
dehydrogenase was found to be decreased in the animals exposed to 1,000
and 2,000 mg/kg DEHP. These results suggest that DEHP can affect
spermatogenesis by altering the activities of the enzymes responsible
for the maturation of spermatozoa. The reduced number of spermatozoa may
be responsible for the antifertilic effects of DEHP.

     Gray and Gangolli (1986) found that DEHP produced seminiferous
tubular atrophy and reductions in seminal vesicle and prostate weight in
4-week-old (but not 15-week-old) rats. Coadministration of testosterone
or gonado-trophins did not protect against DEHP-induced testicular
toxicity, but did partly reverse the depression of seminal vesicle and
prostate weight. Secretion of seminiferous tubule fluid and androgen
binding protein by the Sertoli cells was markedly suppressed within 1 h
of a dose of MEHP, the major metabolite of DEHP, in immature rats. This
occurred less rapidly in mature rats. However, in distribution studies,
^C-MEHP penetrated the blood testis barrier only to a very limited
extent. These findings suggest that DEHP, rather than MEHP, act
initially to cause Sertoli cell injury, the subsequent loss of germ
cells occurring as a consequence of this. Some features of the
testicular lesion could be reproduced in primary cocultures of rat
Sertoli and germ cells. The initial Sertoli cell lesion may explain
testicular toxicity despite the limited extent to which the monoester
can penetrate the blood testis barrier. Three metabolites of MEHP were
much less toxic in culture than MEHP itself, suggesting that MEHP may be
the active testicular toxin from DEHP.

     Sjoberg et al. (198Sc) studied the kinetics of DEHP in immature and
mature rats.  DEHP was not found to any significant extent in the
peripheral plasma after an oral dose of 1,000 mg/kg. High plasma levels
of MEHP were found, with maximal plasma concentrations ranging from 48
to 152 ng/mL.  An in vitro assessment of plasma protein binding indicated
that MEHP was approximately 98% bound in all age groups, and no age-
related difference in the elimination half-life was observed. The amount
of DEHP-derived material excreted in urine was twice as high in 25- as
in SO-day-old rats. The mean area under the curve for MEHP concentration
in plasma vs time was also significantly larger in 25- than in 40- and
60-day-old rats. These observations suggest that the extent of
absorption and/or formation of MEHP from DEHP, and hence total exposure
to MEHP and its metabolites, is higher in young than in more mature rats
after oral administration of DEHP. It is probable that this finding is
relevant to the age-related difference in the toxic effects of DEHP on
the testis.

     Rhodes et al. (1986) reported that oral and intraperitoneal
administration of DEHP to the marmoset monkey at doses up to
5 irunole/kg/day DEHP for 14 days did not induce morphological or
biochemical changes in the tescls comparable with those obtained  in rats
given the same amount of DEHP. This interspecies difference in
testicular effects reflects that seen in hepatic effects and may be
related to the postulated difference Ln metabolism of DEHP between rats
and primates.

-------
                                                 ToxicoLogical Daca    59

      Sjoberg  et al.  (1985c) also  studied  the  testicular  response of DEH?
 In immature and mature  rats and effect on testis. After  14 daily oral
 doses of  1.0  g/kg DEHP  to 25-, 40-. and 60-day-old  rats,  testicular
 damage was observed  in  the youngest age group only.

      Saxena et al. (1985) reported that DEHP produced significant
 histochemical and hlstological alterations  in the testes  of  rats given
 daily oral doses of  2.000 mg/kg DEHP for  7  days. Histological
 examination revealed marked degeneration  of the seminiferous tubules
 Histochemical examination revealed patchy loss in the activity of
 succinic  dehydrogenase, NADH-diaphorase,  and acid phosphatase, and
 increases in adenosine  triphosphate, glucose-6-dehydrogenase, and
 alkaline  phosphatase. This suggests that  inhibition of enzymes involved
 in energy synthesis  may result in cellular  damage to the  target tissue

      Mangham et al.  (1981) administered 2,500 mg/kg/day orally to male
 Wistar rats for 7 or 21 days. DEHP produced a significant reduction in
 the relative weight  of  the testes. Light  microscopic examination of che
 testes of treated animals revealed that 50  to 100% of the germinal cells
 were  absent from the seminiferous tubules. No effects were observed in
 the interstitial cells or on the Sertoli  cells.

      Several studies have shown that in rodents administered DEHP,
 decreases in testicular zinc concentrations and increases in
 testosterone accompany testicular atrophy. Oishi and Hiraga  (1980) fed
 JCL:ICR mice 2,000 ppm DEHP in the diet for 1 week.  Treatment with DEHP
 increased relative testes weight and decreased testicular testosterone
 concentrations.  Oishi and Hiraga (1983) found that young male rats
 orally administered  2.0 mL/kg of DEHP developed testicular atrophy and
 lost  testicular zinc. Coadministration of 2 mL/kg of DEHP and 30 ppm
 zinc  sulfate in the  diet for 10 days did  not result in the prevention of
 testicular atrophy.   Despite increases in  the zinc concentrations in che
 liver  and serum,  testicular zinc concentrations were not  increased  This
 suggests that the testicular effects of DEHP are not related to
 inhibition of gastrointestinal absorption of zinc.

     Oishi (1985) reported that the reversibility of DEHP-induced
 testicular atrophy was limited in the Wistar rat.  Animals were orally
 administered 2,000 mg/kg DEHP for 14 days, and then half of the animals
were sacrificed.  The testicular weights of the rats sacrificed at 14
 days were significantly less than those of the controls.  In addition,
 testicular testosterone and zinc concentrations were reduced. The
 remaining rats were allowed 45 days to recover and then were sacrificed
At the end of the recovery period the serum testosterone  levels in the
 treated rats were comparable to those of controls,  but the testicular
 zinc and testosterone levels were lower than the control values. In
 addition,  testicular weight of the treated animals was decreased and,
histologically,  only a small number of seminiferous tubules showed
 spermatogenesis.

     Gray and Buttervorth (1980) found that DEHP-induced  testicular
atrophy was age  dependent in Uistar rats.  Oral administration of 2,800
mg/kg/day DEHP for 10 days produced tubular atrophy in 4-week-old racs
but had no effect on the testes of 15-week-old rats. In rats that were
 10 weeks old,  testicular weight was not affected,  but histological
examination revealed that 5 to 50% of che tubules were atrophlc compared

-------
60   Section 4

to the controls. In both the 4- and 10-week-old animals,  advanced
germinal cells were lost, with only spermatogonia,  Sertoli cells,  and
occasional primary spermatocytes remaining.  The testicular effects of
DEHP were reversible whether treatment was stopped prior  to,  or
continued until after, the time at which rats normally become sexually
mature.

     In an attempt to further elucidate the age-dependent response of
DEHP, Sjoberg et al. (1986b) treated groups of 25-, 40- and 60-day-old
Sprague-Dawley rats by oral administration with DEHP at dose levels of
1,000 or 1,700 mg/kg/day for 14 days.  Body weight gain was retarded in
all groups, and testicular weight was markedly reduced in the 25-  and
40-day-old rats in the high-dose group. Repeated administration of 500
mg/kg DEHP intravenously (3-h infusions every other day for 11 days;  six
infusions per animal) to 25- and 40-day-old rats did not  cause age-
dependent effects. This suggests that the testicular response of younger
rats is not greater than that of older rats.  It may be that the age-
related difference in testicular response after oral administration is
due to age differences in the pharmacokineti.es of DEHP (Sjoberg et al.
1985c).  However, because the pharmacokinetics of DEHP after intravenous
administration has not been investigated, this possibility cannot be
verified.
     Gray et al. (1982) studied species differences in the testicular
toxicity of DEHP. Groups of Sprague Dawley rats and DSN Syrian hamsters
were orally administered 2,800 or 4,200 mg/kg DEHP by oral intubation
for 9 days. In the rats, DEHP produced severe seminiferous tubular
atrophy. In contrast, the hamster showed only minor changes in response
to DEHP. The rate of intestinal hydrolysis of DEHP was significantly
slower in the hamsters than in the rats, which may account for the
observed species differences. In the same study, MEHP, the major
metabolite of DEHP, did cause focal seminiferous tubular atrophy in
hamsters.

4.3.4.3  Dermal
     Human. • No data were available on the reproductive effects of DEHP
in dermally exposed humans.
     Animal.  No data were available on the reproductive effects of DEHP
in dermally exposed animals.

4.3.4.4  General discussion
     DEHP has been shown to be a reproductive toxicant in both male and
female mice. Its effects were characterized by reduced fertility.
production of fewer litters by the breeding pairs, and the birth of
fewer male and female live pups per litter in the  treated groups than  in
controls. However, DEHP did not affect  libido or mating performance at
dietary doses of up to 3,000 ppm (approximately 390 mg/kg/day).
     At high doses, exposure to DEHP has been demonstrated to cause
testicular atrophy, characterized by degeneration  of  the  seminiferous
tubules, in several species. The testicular effects of DEHP  appear to  be
related both to dose and duration of exposure. Mechanistic studies of
DEHP-induced testicular effects have focused on the role  of  zinc,  which

-------
                                                 TOXLCOLogical Data   61

is normally found at relatively high concentrations in testicular
tissue. DEHP-induced testicular injury is accompanied by a decrease in
the zinc content in the gonads,  and an increase in the urinary excretion
of this element. Although an association between testicular atrophy and
zinc depletion has been demonstrated, it is not known whether a cause-
effect relationship exists, or what role zinc has on this effect. The
age of the animals tested in studies with DEHP has been the focus of
several studies. Oral administration of DEHP at high doses has been
shown to produce testicular atrophy in immature (but not in mature)
rats. Data from metabolism studies suggest that gastrointestinal
absorption of the metabolite MEHP is greater in young animals,
suggesting that larger amounts of this active toxicant reach the target
tissues.

     Numerous studies indicate that orally administered DEHP induces
dose-dependent testicular lesions. Moreover, these lesions have been
shown to be both species and age dependent. The available evidence
indicates that these species- and age-dependent differences may be
related to differences in the pharmacokinetics of DEHP, primarily
through differences in rate of intestinal hydrolysis of DEHP to MEHP
However,  age- and species-dependent differences in the metabolic
conversion of MEHP to glucuronidates and/or oxidation products cannot be
discounted.

4.3.5  Genotoxicity

     DEHP has been extensively tested in short-term genotoxicity assays
due to its reported carcinogenicity and its inclusion in the
collaborative study program of the International Program on Chemical
Safety (Ashby et al.  1985,  Ashby 1986).  The major metabolites of DEHP,
MEPH and 2-ethylhexanol, have also been studied sufficiently to permit a
determination of their genotoxicity. DEHP has not been shown to be
mutagenic in most microbial and mammalian assay systems.  Most of the
data also suggest that MEHP and 2-ethylhexanol are not mutagenic.

4.3.5.1  Genotoxicity studies

     The genotoxicity of DEHP has been studied extensively (Butterworth
1984, CMA 1982,  Douglas et al. 1986, Hopkins 1983, Kozumbo et al. 1982.
Parry et al.  1985,  Phillips et al. 1982, Priston and Dean 1985, Probst
and Hill 1985,  Sanner and Rivedal 1985,  Toraita et al. 1982, Vogel 1985,
Von Halle 1985,  Williams et al.  1985, Wurgler et al. 1985, Yoon et al.
1985, Yoshokawa eC al.  1983, Zeiger and Haworth 1985, Zeiger et al.
1982). An overall summary of the results in specific test systems is
presented in Table 4.2.

4.3.5.2  Discussion

     The overall weight of evidence indicates that DEHP is not mutagenic
in microbial or in in vivo and in vitro mammalian test systems. The
available data have not shown evidence of direct damage by DEHP to DNA
or chromosomes.  Although there are several equivocally positive studies.
most of the evidence suggests that MEHP is not genotoxic. The database
for 2-ethylhexanol is less extensive, but the results suggest that it  is
not mutagenic.

-------
62     Seccion  <4
                                   Tabte 4.2. Results of short-twin assays
                                         OEHP
                                                                 MEHP
          Description of test
  Positive    Negative      Positive    Negative
                                    2-Ethylhexanol

                                  Positive   Negative
    Bacteria
      Salmonella mutagemcity
      £ co/i mutagemcity
      B subnlu  DNA repair

    Yeast
      Sacctiaromyces cerevisiae
        Mutation
        Crossing-over
        Gene conversion
        Aneuploidy
      Schuosaccharomycti pombt
        Mutation
      Aiptrgilliu
        Crossing-over
        Aneuploidy
    In fitro i
    geootoxlciry
     Chromosomal damage
     Sister chromatid exchanges
     Polyploidy
     Aneuploidy
     Mitotic spindle
     Unscheduled DNA synthesis
     DNA damage
    la ritro
     Mouse lymphoma
     Mouse Balb/c3T3
     Chinese hamster ovary
     Chinese hamster V79
     Human lymphoblasu TK
     Human lymphoblasu AHH
   to fifoi
   genotoxktty
     Micronucleus test
     Human lymphocyte
       chromosome damage
     Sperm morphology-mouse
     Sperm morphology-rat
     Rat bone marrow
       chromosome damage
     Syrian hamster traaspla-
       cental chromosome damage
     Body I\w4»/Salmonella
     Unscheduled DNA synthesis
     DNA damage
     Dominant lethality in mice'
 \"
 0
 0
 0
 0
 0
 0
 0

 I
 K')'
 0
0
0
0
0
0
0
0

0
0
0

I

0
0
1C)
I
16
 2
 2
17
 7
 3
 0
 0
 8
 S
                           0
                           0
                           0
                           0
             0
             0
2
I
0
0
0
0
0
            0
            0
            0
            2
            0
            0
            0
            0

            0
            0
            0

            I

            0

            ll
            0
                      0
                      0
                      0
                      0
          0
          0
0
I
0
0
0
2
I
         0
         I
         1m
         0
         0
          I
         0

         0
         0
          1

         0

         0
                       Im"
                      0
                      0
                      0
                      0
             0
             0
1m
0
0
0
0
0
0
            0
            0
            0
            0
            0
            0
            0
            0

            0
            0
            0

            0

            I

            l(')
            0
                        0
                        0
                        0
                        0
0
0
0
0
0
1
0
           I
           0
           0
           0
           0
           0
           I
           0

           0
           0
           I

           0

           0

           2
           1

-------
                                                                        ToxicoLogLcaL  Daca     63
                                        Table 4.2 (cootiooed)
DEHP
Description of test
In >itT« niainnulUfl
cell transformation
Syrian hamster embryo
Mouse C3H
Mouse BALB/e 3T3
Virus-enhanced
transformation
Virus-enhanced survival
Drotofkila
Sex-linked recessive
lethal
Recombination
Somatic mutations/deletions
Recombination/somatic
mutation/aneuploidy
Test of promotion In rltro
Metabolic cooperation
Anchorage independence
Promotion of cell
transformation
Hepatocellular
hyperplastic foci
Mouse
Rats
Skin painting
SENCAR mouse
CD-I mouse
Positive


3
1
0
1

1

0

0
1m
0


2 + 1m
1
0



1
0

1
0
Negative


0
1
3
0

0

1

|
0
1


1
0
1



0
3

0
1
MEHP
Positive


1
0
0
0

0

0

0
0
0


0
1
0



0
0

0
0
Negative


0
1
1
0

0

0

0
0
0


0
0
1



0
0

0
0
2-Ethylhexanol
Positive


0
0
0
0

0

0

0
0
0


0
0
0



0
0

0
0
Negative


2
0
1
0

0

0

0
0
2


0
1
0



0
0

0
0
   "Numeric values are number of studies.
    m — marginal.
   e(') ~ equivocal.
    i  — inconclusive.
   'The positive result seen in this dominant lethal assay has not been clarified as being induced by a genetic
effect  or a developmental effect caused by a nonchromosomal mechanism of action.

   Source  Modification (update) of table presented in CPSC 1985.

-------
 64    SecCion  4

 4.3.6  Carcinogenicity

      DEHP  is  a  probable human carcinogen, classified as weight-of-
 evidence Group  B2 under the EPA Guidelines for Carcinogen Risk
 Assessment (EPA 1986a). Evidence on potential carcinogenicity from
 animal  studies  is "Sufficient," and the evidence from human studies is
 "No  Data"  (EPA  1986b).

 4.3.6.1  Inhalation

      Human.   No data were available on the carcinogenic effects of DEHP
 in humans  exposed via inhalation.

      Animal.  No data were available on the carcinogenic effects of DEHP
 in animals  exposed via inhalation.

 4.3.6.2  Oral

      Human.   No data were available on the carcinogenic effects of DEHP
 in orally  exposed humans.

      Animal.  The carcinogenicity of DEHP has been tested in a bioassay
 using Fischer 344 rats and B6C3F1 mice (NTP 1982. Kluwe et al. 1982a,b)
 Groups of  50  rats per sex were fed diets that contained 0 (control),
 6,000, or  12,000 ppm DEHP, and groups of 50 mice per sex were fed diets
 that  contained  0 (control), 3,000, or 6,000 ppm DEHP (purity >95.5%) for
 103 weeks.   A  1-  to 2-week nontreatment observation period was allowed
 after the  termination of exposure, after which all survivors were
 sacrificed  and  examined both grossly and microscopically.
 Histopathological examinations were conducted on all animals sacrificed
 or discovered dead (if not precluded by autolysis).  The tissues examined
 included skin,  lungs, bronchi, trachea, larynx, bones,  bone marrow,
 spleen, lymph nodes, heart, liver, pancreas,  esophagus, stomach, small
 intestine,   large intestine, kidney, bladder,  pituitary, adrenal, thymus.
 thyroid, parathyroid, salivary gland, mammary gland, testis or ovary,
 prostate and  seminal vesicles or uterus, and brain.

     A dose-related decrease in body weight gain was observed in the
male  rats  throughout the study.  Body weight gain was also reduced in che
high-dose  females. Daily mean food consumption was slightly decreased in
 the low- and high-dose rats of both sexes when compared to the controls
No other compound-related clinical signs of toxicity were reported  No
 statistically significant dose-related trends in mortality were
observed.  In male rats, 60% of the control, 56% of the low-dose, and 66%
of the high-dose animals lived to the end of the study. In female rats.
 72% of the  control.  68% of the low-dose, and 76% of the high-dose
animals were alive at the end of the study. A sufficient number of racs
were alive  at the end of the study to be at risk from late-developing
 tumors.

     The incidence of female rats wich hepatocellular carcinomas at the
higher dose was  greater than that in controls by pairwise comparisons,
and there was a  significant (P < 0 05) dose-related trend. A dose-
 related trend was also observed for hepatocellular carcinomas in the
males, but  pairwise comparisons did not indicate a statistically
significant increase.

-------
                                                 Toxicological Data   65

     A summary of the incidence of hepatocellular carcinomas and
neoplastic nodules observed in the rats is presented in Table 4.3.

     The incidence of female rats with neoplastic nodules was greater
for the higher-dose group than in the controls and displayed a
significant (P < 0.05} dose-related trend. The incidence of male rats
with neoplascic nodules was not significantly increased when evaluated
by either pairwise comparisons or trend tests  When combined, the
incidence of rats with either hepatocellular carcinomas or neoplastic
nodules exhibited a significant (P < 0.01) dose-related trend in both
sexes, and was significantly (P < 0.05) greater than the controls for
female rats at both doses and for male rats at the higher dose level by
pairwise comparisons.

     The incidences of male rats with pituitary tumors, thyroid C-cell
tumors, or testicular interstitial-cell tumors were all significantly
(P < 0.05) reduced in treated animals by pairwise comparisons or by
trend test. Myeloraonocytic leukemia, mammary gland fibroadenoma,
clitoral gland carcinoma, and uterine endometrial stroraal polyps were
also observed in one or more rats in this study,  but their incidences in
treated animals did not differ significantly from chose in the controls.

     In mice,  there was a dose-related decrease in the body weights of
the female from week 25 to the end of the study.  Daily mean food
consumption was similar in the treated and control groups throughout the
study. No other compound-related clinical signs of toxicity were
reported. In male mice, 68% of the control, 76% of the low-dose, and 70%
of the high-dose animals lived to the end of the study. In female mice,
72% of the control, 68% of the low-dose,  and 76% of the high-dose
animals were alive at the end of the study. Low-dose females had
significantly shortened survival compared with that of the controls
(P - 0.006), but no positive trend was observed with respect to survival
when all groups were compared. A sufficient number of mice were alive at
the end of the study to be at risk from late-developing tumors.

     There was a statistically significant increase in the incidence of
hepatocellular carcinoma in both the high-dose males and high-dose
females when compared with the controls.  A significant (P < 0.05),
dose-related trend of Increasing hepatocellular carcinoma was also
observed for the alee of both sexes. A summary of the  incidence of
hepatocellular carcinomas and hepatocellular adenomas  observed in the
mice is presented in Table 4.4.

     Metastases of the hepatocellular carcinomas were  observed in the
lungs of 12 treated males and B treated females. The incidence of
neoplastic lesions of other sites in the treated animals did not differ
significantly from that of controls.

     Northrup et al. (1982) have questioned the validity of  the NTP
bioassay of DEHP. They suggested that the MTD was exceeded in the low-
and high-dose female mice, low- and high-dose male rats, and high-dose
female rats because body weight gain was depressed by  more than 10%  in
these groups.  However, Kluwe et ai. (1983) have replied that the 10%
weight differential was only a guideline and that the  primary reason  for
not exceeding the estimated MTD was to avoid excessive early mortality
which might preclude tumor development and to avoid pathological lesions

-------
66    Seccion
                 Table 4.3.  Incidence of hepatocellular carcinomas and
                   neoplastic nodules observed in DEHP-dosed rats"

                  Dose     Hepatocellular    Neoplastic    All liver
                 (ppm)     carcinomas       nodules       tumors

                                      Males

                      0        1/50          2/50        3/50
                  6.000        1/49          5/49        6/49
                 12.000        5/49          7/49       12/49

                                     Females
0
6.000
12.000
0/50
2/49
8/50
0/50
4/49
5/50
0/50
6/49
13/50
                    "References- NTP 1982; Kluwe et al. 1982a,b.
                Table 4.4.  Incidence of hepatocellnlar carcinomas and
                hepatocellular adenomas observed in DEHP-dosed mice"

                Dose     Hepatocellular   Hepatocellular   All liver
                (ppm)     carcinomas       adenomas       tumors

                                      Males
                   0         9/50            6/50         14/50
                3000        14/48            11/48          2S/48
                6000        19/50            10/50         29/50

                                     Females
0
3000
6000
0/50
7/50
17/50
1/50
5/50
1/50
1/50
12/50
18/50
                   "References: NTP 1982: Kluwe et al. 1982a.b.

-------
                                                  Toxicological Data   67

 other than neoplasia (Haseman 1985).  In the case of the DEHP bloassay.
 NTP (1982) concluded that both of these goals had been met. Survival was
 not adversely affected by DEHP,  and statistical analyses failed to
 demonstrate any correlation between the occurrence of nonneoplastlc
 tissue lesions (testicular atrophy In the high-dose male rats and mice
 and pituitary hypertrophy in the high-dose male rats) and the
 development of hepatocellular tumors  in rats and mice of both sexes
 (Kluwe et al.  1982a,b,  1983).  However,  there is a second issue with
 regard to the use of the  MTD in  this  study.  As discussed below,  there is
 some evidence that suggests that peroxisorae  proliferation,  which occurs
 in both mice and rats at  the dose levels  used in the NTP bioassay,  is
 involved in a secondary mechanism of  cancer  induction (Reddy et  al.
 1986).  If this hypothesis is correct,  it  suggests that at low doses,
 where  peroxisome proliferation does not occur,  one would not expect
 cancer to develop (Turnbull and  Rodricks  1985,  Warren et al.  1982).

     Northrup  et al.  (1982)  also questioned  the significance of  the
 DEHP-induced increase in  liver tumors  in  the  NTP bioassay because  of
 intralaboratory variations  in  incidences  of  liver tumors in controls  in
 bioassays of guar gum,  butyl benzyl phthalate,  and di(2-ethylhexyl)
 adipate,  which were  conducted  simultaneously  in the same rooms as  the
 DEHP study.  They reported that statistical analysis indicated that  there
 was  a  significantly  lower incidence of  tumor-bearing animals  among  the
 female mice  used in  the DEHP control group.  In  addition,  they suggest
 that the  number of tumor-bearing male or  female mice in the DEHP
 treatment groups  did  not  substantially  differ  from the  average for  all
 pooled controls.  They also  report  that  in male  rats,  the incidence  of
 tumor-bearing  animals had a  highly negative dose  response (P  < 0.001),
 and  female rats had a marginally positive dose  response (P  -  0.045)
 when paired  to  the pooled controls. However, Kluwe  et al. (1983)
 reported  that  there was relatively little variation in  liver  tumor
 incidences among both NTP historical controls and  concurrent  laboratory
 controls, and  that the data  reported in the concurrent  study  controls
 were quite similar to the historical controls (Haseman  1983). In
 addition, Kluwe et al.  (1983)  reported  that replacement of  the
 concurrent study controls with either concurrent laboratory controls  or
 pooled controls from bioassays performed at approximately the same  time
 did not alter the statistical significance of the DEHP  effects.
 Replacement of the concurrent study controls with historical  controls
 strengthened the statistical significance of the difference in liver
 tumor incidence.

     Carcinogenic effects were not reported in  2-year bioassays  with
 Sherman or Wistar rats of both sexes fed diets which contained up to
 5,000 ppa DEHP (Carpenter et al.  1953, Harris et al. 1956). However,  the
design and reporting of these studies  were not adequate  to  fulfill  the
stringent Good Laboratory Practices and Good Management  Practices
 required of today's toxicity testing guidelines.

4.3.6.3  Dermal

     Human.  No data were  available on the carcinogenic effects  of DEHP
in dermally exposed humans.

-------
68   Section 4

     Animal.  No data were available of the carcinogenic effects of DEHP
In dermally exposed animals except for those reported in Sect. 4.3.6 4.
The carcinogenic risk due to dermal exposure to DEHP is likely to be
low, based on observations of poor dermal absorption (El Sisi et al.
1985) and an absence of initiation or promotion activity in mouse skin
painting studies (Ward et al. 1986).

4.3.6.4  Possible mechanisms of carcinogenesls

     Several initiation-promotion studies have been performed with DEHP
Ward and coworkers (Diwan et al. 1983, 1985; Ward 1983, 1986) conducted
skin-painting studies to assess the promotional activity of DEHP. Single
topical applications of 50 pg of 7,12-dimethylbenz(a)anthracane (DMBA),
an initiator, were applied to the skin of CD-I mice, followed by DEHP
(98.1 pg in acetone) twice weekly for 40 weeks. A known promoter, 12-0-
tetradecanoyl-phorbol-13-acetate (TPA),  was used as a positive control
Under the conditions of this study, DEHP was not a promoter. No tumors
were observed in animals treated with DMBA followed by DEHP. In
contrast, 97% of the mice treated with DMBA followed by TPA developed
skin tumors.

     In a second study performed by Ward and coworkers, female SENCAR
mice received a single topical application of 20 /jg of DMBA, followed by
2 pg of TPA twice weekly for 2 weeks, followed by 100 /jg of DEHP twice
weekly. Animals treated with TPA, mezerein, or acetone served as
controls. Under the conditions of the study, DEHP showed some late
promotional activity, producing 6.4 papillomas per mouse. In contrast,
DMBA + TPA + mezerein produced 23 papillomas per mouse, DMBA + TPA + TPA
gave 26.4 papillomas per mouse, DMBA + DEHP + DEHP gave 0.9 papillomas
per mouse, and DMBA alone produced no tumors.

     Albro et al. (1982) reported that radioactivity from carbonyl-
labeled DEHP did not bind to purified protein, RNA, or DNA from rat
liver in vivo. 0-ethyl-(l-14C)-hexyl-labeled DEHP and MEHP did appear to
bind strongly with purified DNA, but the label from free 14C-labeled 2-
ethylhexanol did not. The apparent binding of DEHP and MEHP was not
exchangeable and was not demonstrated to be covalent.

     DeAngelo et al. (1985a) examined the protein kinase activity of
isolated plasma membranes from the livers of rats treated with three
promoting regimens. Male rats first received either an initiating dose
(30 rag/kg) of the hepatocarcinogen diethylnitrosamine or the 0.9% NaCl
solution vehicle by intraperitoneal injection at 18 h following partial
hepatectomy (to induce cell proliferation). Ten days later, the three
promoting regimens were begun. These consisted of 10 weeks of treatment
with either (a) a choline-deficient (CD) diet, (b) a choline-
supplemented (CS) diet containing 0.06% phenobarbital (PHB) (CS plus
PHB),  or (c) a CD diet containing 0.06% PHB (CD plus PHB). In addition,
two other groups of rats received either (a) a CS diet containing 2%
DEHP (CS plus DEHP) or (b) a CD diet containing 2% DEHP  (CD plus DEHP)
DEHP had been shown previously to inhibit the development of putative
preneoplastic gamma-glutamyl transpeptldase (GOT) positive foci in  rat
liver.  Total liver plasma membrane protein kinase (PK) activity, using
both protamine sulfate andhistone. was cyclic adenosine 3':5'-
monophosphate independent and did not appear to be a marker of

-------
                                                  ToxLcologicaL  Daca    69

 promotion.  Protein kinase  activity, however,  was  increased  by both DEHP
 (which suppresses  the  development of GGT  positive foci)  and a CD  diet
 (which promotes  the appearance of GGT positive  foci).  The CD, CS  plus
 PHB,  and CD plus PHB dietary  regimens, which  promote  the appearance of
 GGT positive foci,  induced the phosphorylation  of a 40,000  molecular
 weight (raw)  plasma membrane protein in vitro  by endogenous  protein
 kinases.  Plasma membranes  from DEHP-treated rats  did  not demonstrate
 phosphorylation of this 40,000-mw protein. DEHP dietary  treatment also
 blocked the  ability of the epidermal growth factor to  enhance the
 phosphorylation of its 175.000-mw receptor protein in  isolated  liver
 plasma membranes.  These results suggest that  the  phosphorylation of a
 40,000-raw plasma membrane  protein may be  important to  the early
 promotional  phase  of liver carcinogenesis, and  that one  mechanism by
 which  DEHP  inhibits the emergence of GGT  positive  foci may  be by
 blocking the  response  of initiated cells  to stimulation  by  epidermal
 growth factor.

     As  discussed  above, DEHP has been demonstrated to cause an increase
 in  the incidence of liver  tumors in the F344  rat  and  the B6C3F1 mouse
 (NTP 1982).  The weight of evidence suggests that  DEHP  is not genotoxic
 in  either in vitro  or  in vivo assay systems.  Several hypotheses have
 been proposed to explain the mechanism(s) by  which DEHP  induces liver
 tumors  in rodents.   Reddy and Lalwani (1983) suggested  that  DEHP belongs
 to  a novel class of nonmutagenic chemical carcinogens which alter the
 level  and function  of  peroxisomes in rodent liver  cells. This hypothesis
 assumes  that the hepatocarcinogenesis of  DEHP is not related to a direct
 initiating effect of DEHP  (or its metabolites), but is linked to
 metabolic disturbances emanating from a sustained  increase  in the number
 of  peroxisomes in  liver cells (Rao and Reddy  1987). The  terra "peroxisome
 proliferation" refers  to an increase in the number of subcellular
 organelles called peroxisomes in response to  administration of a series
 of  compounds collectively called "peroxisome proliferators" (CPSC 1985)
 The peroxisome proliferating compounds are structurally  diverse. The
 only thing these compounds have in common is  the capability to induce
 increased numbers of peroxisomes and to produce hypolipidemia. They have
 been characterized by  the CPSC (1985)  as having the following features

 • Induction of increased numbers of peroxisomes in several  species,

 • Induction of variable degrees of hepatocyte hyperplasia,

 • Lowering of the concentrations of plasma lipids,

 • Induction of liver carcinogenesis,  and

 • Weak or absent mutagenicity in in vitro rautagenicity bioassays.

     The phenomenon of peroxisome proliferation and its  relationship co
 the carcinogenicity of DEHP has been studied extensively and has been
 the subject of several recent reviews  (Reddy et al. 1986, Rao and Reddy
 1987, CPSC 1985).

     Liver enlargement occurs within a few days of dietary
administration of peroxisome proliferators such as DEHP, rapidly reaches
a steady-state level (in 7 to 10 days),  and continues as long as
administration of the proliferator continues  (Reddy and  Lalwani 1983)
The peroxisome proliferators are not equipotent in the degree of

-------
70   Section 4

observed hepatomegalic effect chey induce.  DEHP  is  a relatively weak
peroxisome proliferates when compared to  compounds  such  as  ciprofibrate
(by a factor of 1.000) (Reddy et al.  1986).  The  liver enlargement
associated with the administration of peroxisome proliferators  is  due  to
both hyperplasia and hypertrophy of hepatocytes  (Rao and Reddy  1987)
The hyperplasia is evident as early as 24 h after treatment,  reaching  a
maximum in 5 to 7 days. Hypertrophy of the hepatic  cells is secondary  co
marked increases in the number and volume of peroxisomes.
     The development of hepatocellular carcinomas due to administration
of a peroxisome proliferator was first reported  by  Reddy et al.  (1976)
Since then, several hypolipidemic compounds and  plasticizers  that  are
peroxisome proliferators have been shown  to induce  liver tumors in mice
and rats (Rao and Reddy 1987).  The latency period and incidence of
tumors appear to correlate well with the  ability to induce  peroxisome
proliferation. Potent peroxisome proliferators such as Wy-14643,
ciprofibrate, and methyl clofenapate induce liver tumors in nearly 100%
of treated rats after SO to 60 weeks of administration (Rao and Reddy
1987).  Less potent peroxisome proliferators such as clofibrate  and DEHP
induce liver tumors after 70 and 104 weeks,  respectively (Svoboda  and
Azarnoff 1979, NTP 1982). The majority of the liver tumors  induced by
the peroxisome proliferators are well-differentiated hepatocellular
carcinomas, with predominantly trabecular histological patterns (Rao and
Reddy 1987). Metastases are encountered in 20 to 40% of rats and mice
with peroxisome-proliferator-induced hepatocellular carcinomas  (Rao and
Reddy 1987).
     The lack of mutagenicity of the peroxisome  proliferators in in
vitro and  in vivo assays has generated interest  in the mechanism of
action by which these compounds induce liver tumors. Several hypotheses
have been posed stating that the carcinogenicity of these compounds is
due to biologically active products of the proliferated peroxisomes
rather than direct DNA damage. The evidence supporting this hypothesis.
which was  recently summarized by Rao and Reddy  (1987), includes (1) a
consistent association between the induction of peroxisome proliferation
and liver carcinogenesis; (2) sustained and specific induction of H202-
producing beta-oxidation enzymes;  (3) increased intracellular levels of
H202 in livers of rats with peroxisome proliferation; (4)  increases of
hepatic lipofuscin and increased hepatic levels of diene conjugates,
indicative of increased lipid proliferation; and (5) marked  inhibition
of peroxisome proliferator-induced hepatocarcinogenesis by antioxidants
     The importance of oxidative stress on  the  process of  initiation and
promotion  of carcinogenesis  remains unknown. The peroxisomes contain a
variety of enzymes associated with  llpid metabolism  (CPSC  1985).  Several
oxidation  reactions can be catalyzed by  the enzymes  residing in the
peroxisomes. These reactions employ as their terminal oxidase  an  H202-
generating flavin oxidase. The H202 produced is  converted  to H20  by
catalase,  either by directly converting  H202 to H20  plus 1/2 02 or  by
utilizing  two hydrogen atoms  from  suitable  molecular donors  to  convert
H202 to two molecules of H20 plus  an  oxidized substrate.

     The administration  of drugs  such  as  hypolipidemic  agents  and
several phthalates results  in  a  sudden proliferation of peroxisomes
followed by  a significant  increase in  parcicle-bound catalase  activity

-------
                                                  Toxicological Data   7L

 The mechanism by which peroxisome proliferators exert their effects is
 not well understood at present and has been the subject of considerable
 research. Several investigators have proposed that these compounds
 induce peroxisome proliferation by increasing the influx of lipids, such
 as long-chain fatty acids, into hepatocytes (Reddy and Lalwani 1983.
 Havel and Kane 1973).  This results in an increase in the number of
 peroxisoraes, presumably due to the induction and increased synthesis of
 the enzymes and other  components that constitute these organelles (CPSC
 1985). This hypothesis is supported by the finding that feeding rats a
 diet high in fats results in a slight increase in the number of
 peroxisomes (CPSC 1985).  However,  this increase is very slight compared
 to the increase following administration of peroxisome proliferating
 compounds.  The results of several  studies suggest that there is no
 direct lipolytic effect by peroxisome proliferators.  Lake  et al.  (1983)
 reported that in a comparison of the  effects of several phthalate
 esters,  both DEHP and  mono-n-octyl phthalate had hypolipidemic effects,
 while  peroxisome proliferation was seen only with DEHP and its
 metabolites.  Similar findings were reported in a study by  Lazarow and
 DeDuve (1976)  with bezafibrate and clofibrate.  in which both compounds
 had hypolipidemic effects,  but only clofibrate induced peroxisomes.

     An  alternative hypothesis is  that peroxisome  proliferators act as
 substrates  for the  peroxisomal beta-oxidation system  (Reddy and Lalwani
 1983,  Reddy  1987).  This hypothesis is supported  by the fact that  some
 components of  the molecular structure of  the peroxisome  proliferators
 appear as suitable  candidates  for  metabolism by  the peroxisomal enzymes
 (CPSC  1985). The  combined  effect of fatty acid influx with influx of the
 peroxisorae proliferator is  sufficient to  result  in peroxisome
 proliferation.  Phthalate esters appear  to undergo  beta oxidation,  and
 this could occur  in the peroxisomes.  A  study by  Lhuguenot  et al.  (1985)
 suggests that  at  high  doses, MEHP  is  partly  metabolized  by peroxisomes
 A.  M.  Mitchell  et al.  (1985)  reported that peroxisome proliferation  is
 associated with concomitant  increase  of a cytochrome  P-450 species  that
 is  associated  with omega oxidation  of lauric acid  and other  long-chain
 fatty  acids. They proposed  that peroxisome proliferation and cytochrome
 P-450  induction are attempts by the rat hepatocyte to cope with a
 disturbed lipid metabolism  (CPSC 1985). Because of the inability  of  the
 rat to conjugate  these compounds,  their presence leads to  a substrate
 load that is alleviated by  the creation of these alternative pathways
 (CPSC  1985). At high doses of DEHP. excess H202 or other oxygen species
 are produced in excessive amounts because catalase production  does not
 increase as rapidly as  peroxide production.

     Recently, Rao and Reddy (1987) have  proposed that peroxisome
 proliferacors exert their effect by a ligand-receptor-mediated
 mechanism.  According to this hypothesis,  the peroxisome proliferators
 interact with a receptor molecule present in the cytoplasm or  plasma
 membrane of hepatocytes. The formation of a receptor-peroxisome
 proliferator complex results in the generation of a message that  is
 transmitted to the cell nucleus, initiating the process for production
of peroxisomal components (CPSC 1985). The interaction between  the
 ligand and the receptor results in a measurable effect only after a
sufficient number of receptors are  occupied by the ligand.  Rao  and Reddy
 (1987)  have  stated that indirect evidence in favor of this hypothesis

-------
72   Section 6

includes (1) tissue-specific predictable biological response,  (2)
response of extrahepatic hepatocytes to the inductive effects  of these
compounds, (3) inducibility of peroxisome proliferation in hepatocytes
in primary cultures maintained in defined medium,  (4) induction of
specific changes in protein composition in the livers of rats  treated
with peroxisome proliferators which differ in chemical structure,  (5) a
rapid and significant increase in the rate of synthesis of mRNAs for
peroxisomal beta-oxidation enzymes in the liver,  (6)  the rapidity  of
transcriptional response of these genes, and (7)  detection of  a specific
binding moiety in liver cytosol for nafenopin, a  clofibrate-like
peroxisome proliferator.  In a study published by  Lalwani et al -(1983),
3H-nafenopin, a known inducer of liver peroxisomal enzymes, was shown to
bind to a specific, saturable pool of binding sites in rat liver and rat
kidney cortex cytosols. Clifibrate and ciprofibrate,  which are
structurally similar to nafenopin, competitively  inhibited the specific
binding of 3H-nafenopin.  Phenobarbital (a noninducer  of peroxisomes),
4-chloro-6-(2,3-xylidino)-2-pyrimidinylthio acetic acid, and 4-chloro-
6-(2,3-xylidino)-2-pyrimidinylthio (Af-beta-hydroxyethyl) acetamide did
not compete for the specific 3H-nafenopin binding sites. This  study
provides some evidence for the presence of such a receptor. Additional
studies are being conducted to identify and purify the putative receptor
protein.

     DEHP was identified as a peroxisome proliferator in 1976  (Reddy et
al.  1976). DEHP induces a stronger response in the rat compared to the
hamster (Turnbull and Rodricks 1985, Rodricks and Turnbull 1987).  In a
report by the Council European of the Federations of the Industry
Chemical (CEFIC) (1982) presented to the CPSC (1985), it was reported
that the induction of peroxisome proliferation occurs in the marmoset to
a much lower degree than in the rat. However, in the  same report,  it was
also stated that the extent of absorption of DEHP in the marmoset  was
much lower than in the rat. Peroxisome proliferation, such as  that seen
in the rodent, has not been quantitatively demonstrated in higher
primates or in humans (De La Iglesai et al. 1982,  Staubli and  Hess 1975,
Sternlieb 1979). Peroxisome nucleoids are absent from the liver of
humans, are rudimentary in some primates, and are well developed in the
liver of rodents (De La Iglesia et al. 1982). Differences in the
relative proportions of metabolites of DEHP have been described for the
rat, nonhuman primates, and humans. Relatively high quantities of
omega-1 oxidation products of MEHP are formed in primates and humans
when compared to the rat. These metabolites are excreted largely as
conjugated products in humans, primates, and the mouse, but not in the
rat (Albro et al. 1981, 1982). The differences between  the species  in
terms of metabolite profiles and conjugation pathways preclude
definitive conclusions as to the expected sensitivity to DEHP
carcinogenicity (CPSC 1985). Most of the available evidence does suggest
that primates in general are less sensitive to the induction of
peroxisome proliferation than are rodents (Lake et al.  1984).

     Although the absence of mutagenic activity is consistent with other
evidence, which suggests that DEHP exerts its carcinogenic effect by  an
epigenetic mechanism, there are not sufficient data  to  allow a
determination of a dose of DEHP below which peroxisome  proliferation
should not be expected to occur in a long-term study  (CPSC 1985). All  of

-------
                                                   Toxicological Daca   73

  nr«MfM          °f ^ mechanisffl of induction of peroxisome
  proliferation suggest an effect will occur only after sufficient
  concentrations of the inducing compound are reached  The available
  studies (CMA 1984;  1985a.b.C.d; 1986a,b.c(d;  Morton I™?;  Red^y 1985)

  Torecfj rF-^'f^0", ^'^ f™  b"n "° ^"ft""-" «udl.. that  allow
  a precise definition of the relationship between DEHP dose and
  peroxisome proliferation.  In addition,  although there is much indirect
  evidence  relating peroxisome proliferation to carcinogenesis.  this
  Uln-M " rt* T   ?6n Pr°Ved directlv-  Uncil ^ch evidence becomes
  available,  the theories regarding the mechanism of  action  of DEHP-
  induced carcinogenicity must be regarded as plausible but  unproven
  Continuing research  is  necessary to  resolve this  issue.

       Lake  et  al.  (1984)  administered 1,000 mg/kg/day  DEHP  by gavage
  daily to male  Sprague-Dawley rats for 14  days.  Concurrent  control
  TP ITnSn^C?      °nly,Che Corn  o11 vehicl.. DEHP  produced  significant
  (P <  0.01)  increases  in  relative liver weight,  marked increases  in the
  hepatic activities of cyanide-insensitive palmitoyl-CoA  oxidation  a
  specific peroxisomal marker  enzyme,  carnitine  acetyltransferase  (an
  enzyme  located in mitochondrial, peroxisomal.   and microsomal fractions)
  and both total (mitochondrial and peroxisomal)  and heat  labile
  (peroxisomal) enoyl-CoA hydratase activities.   In addition  DEHP
  treatment resulted in inhibition of D-amino acid oxidase activity.  The
 effect of DEHP on hepatic microsoraal mixed-function oxidase parameters
 was also investigated. DEHP markedly induced the combined omega- and
 omega-l-hydroxylation of lauric acid, with smaller increases observed in
 the activities of ethylmorphine tf-demethylase  and 7-ethoxycouraarin  O-
 deethylase. In contrast, the activity of 7-ethoxyresorufin O-deethylase
 was significantly inhibited. DEHP also  induced cytochrome P-450 content
 and produced spectral changes in the properties of hepatic microsomal
 hemoproteins.  Histological  studies revealed marked peroxisome
 proliferation in liver sections from treated rats.

      DeAngelo et al.  (1985b) studied the effect of diets containing 0.1
 0.5.  1.0.  and 2.0% DEHP as  equimolar amounts of its  two major
 metabolites.  MEHP and 2-ethylhexanol. on the induction of liver
 peroxisomes and pre-induced GGT positive (GGT+) foci  GCT+  foci were
 initiated  in the  livers  of  Sprague-Dawley male rats  with a  single dose
 of di.thylnleroM.in.  (DEN)  following partial  hepatectomy.  Controls were
 fed a  semipurified diet  for  10 weeks; experimental groups received  the
 semipurified diet containing the respective compounds.  Induction of
 /rr^lfOBe  Proliferaclon  w« monitored by carnitine  acetyl-transferase
 (CAT)  levels. The 2-ethylhexanol produced essentially  no effect with
 nrSrd ?u^b?r  °f f°Cl> Peroxiso«"e  proliferation,  or liver  weight.
 DEHP and MEHP  induced significant peroxisome proliferation  and
 hepatomegaly.

     Toraaszewski  et al.  (1986)  investigated the hypothesis  that
 hepatocarcinogenesis resulting  from treatment of rats  and mice  with
 peroxisome proliferators  is  linked to increased cellular  levels of
hydrogen peroxide from peroxisomal beta-oxidation. Hale  F344  rats and
 female B6C3F1 nice were treated  for 14 days with DEHP  di(2-
ethylhexyl)adipate (DEHA). or nafenopln.  a hypolipidemic  drug.
Activities of enzymes responsible for the production  [peroxisomal
palmitoyl CoA oxidase (PCO)] and degradation [catalase  (Cat)  and

-------
74   Section 4

glutathione peroxidase  (GSHPx)] of H202 were assayed in liver
homogenaces prepared from created animals. The activities of the
peroxisomal enzymes PCO and Cat were enhanced 5- to 25-fold and 1.5-  to
3-fold, respectively, by treatment with DEHP, DEHA, and nafenopin.  The
activity of GSHPx, a cytoplasmic enzyme, was decreased 40 to 60% in
liver homogenates prepared from treated animals compared to control
animals. A kinetic analysis of the rates of formation of hydrogen
peroxide by PCO, and of degradation of hydrogen peroxide by catalase,
was used to estimate steady-state hydrogen peroxide concentrations
[(H202)] during peroxisomal oxidation of palmitoyl CoA.  Increases in
peroxisomal steady state (H202) for the F344 rat liver homogenates
correlated well with the carcinogenic potential of these chemicals,
determined in previous carcinogenicity studies. Increases in the steady
state (H202) were also calculated for liver homogenates prepared from
mice treated with these compounds. Decreases in liver lipid peroxidation
were observed after treatment with each chemical in both species. The
results of these studies are consistent with an involvement of increased
peroxisomal hydrogen peroxide in the hepatocarcinogenesis of these
compounds.

     Lake et al. (1986) investigated the hepatic effects of DEHP on
peroxisomal and microsomal enzyme activities in the intact animal and in
primary hepatocyte cultures. In the in vivo studies, DEHP produced  large
increases in liver size and peroxisomal enzyme activities in Sprague-
Dawley rats and Chinese hamsters, but had less effect in Syrian
hamsters. These effects were largely reproduced in vitro, where good
responses were obtained with rat and Chinese hamster hepatocytes, but
either little or no effect was observed with Syrian hamster and Dunkin-
Hartley guinea pig hepatocytes. These results demonstrate a good
relationship between the in vivo and in vitro effects of DEHP and
indicate that hepatocyte cultures may be useful for further studies on
the hepatic effects of DEHP on peroxisome proliferation.

     DeAngelo et al. (1986) studied the ability of DEHP to suppress the
development of rat liver preneoplastic lesions. GGT+ foci were initiated
in the livers of Sprague-Dawley male rats with a single dose of
diethylnitrosaraine (DEN) following partial hepatectomy.  Promotion of
foci was induced by feeding the rats a choline-deficient diet (CD).
Control rats were fed a choline-supplemented diet (CS).  The ability of
DEHP to suppress the emergence of GGT+ foci was evaluated by feeding
groups of rats the CD diet containing either 0.1. 0.5. 1.0, or 2.0%
DEHP. The CD diet promoted the number of GGT+ foci above levels  in
control livers. Inclusion of DEHP in the CD diet at levels of 0.5.  1.0,
and 2.0% effectively inhibited the appearance of the foci. However, DEHP
was unable to inhibit the promoting effect of the CD diet at a
concentration of 0.1%. DEHP's ability to block development of GGT+ foci
correlated with its ability to increase liver weight and to induce
carnitine acetyltransferase, a marker of peroxisome proliferation.

     A. M.  Mitchell et al. (1985) used a primary rat hepatocyte  culture
system to determine the proximate peroxisome proliferator(s) derived
from DEHP.  DEHP was administered orally to rats, and the urinary
metabolites were isolated and identified  The major metabolites  were
those resulting from initial omega- or omega-1-carbon oxidation  of the

-------
                                                  TOXI.COLogical Data   75

 mono(2-ethylhexyl) moiety. These metabolites,  together with MEHP and 2-
 ethylhexanol, were added to primary rat hepatocyte cultures, and the
 effect on peroxisomal enzyme activity was determined.  The omega-carbon
 oxidation products (mono(3-carboxy-2-ethyl-propyl) phthalate and
 mono(5-carboxy-2-ethylpentyl) phthalate]  and 2-ethylhexanol produced
 little or no effect on cyanide-insensitive palmitoyl-CoA oxidation, a
 peroxisomal marker. Mono(2-ethyl-hexyl) phthalate and the omega-1-carbon
 oxidation products [mono-(2-ethyl-5-oxohexyl)  phthalate and mono(2-
 ethyl-5-hydroxyhexyl) phthalate]  produced a large (7-  to 11-fold)
 induction of peroxisomal enzyme  activity.  Similar structure-activity
 relationships were observed for  the induction  of cytochrome P-450-
 mediated lauric acid hydroxylase  and the  increase in cellular coenzyrae A
 content.  In contrast. A. M.  Mitchell et al.  (1985)  reported that oral
 administration of MEHP (150 or 250  mg/kg)  to male guinea pigs did noc
 produce hepatic peroxisome proliferation.  Addition of  MEHP (0 to 0.5 mM)
 or one of the active  proliferators  in the  rat  to primary guinea pig
 hepatocyte cultures also failed  to  produce an  induction of peroxisomal
 beta oxidation.

      Kornbrust et al.  (1984)  performed experiments  to  test the
 hypothesis that the hepatocarcinogenicity  of DEHP is due to its ability
 to produce DNA damage,  either directly or  as a result  of the
 proliferation of peroxisomes  and  accompanying  increased production  of
 H202 and  other DNA-damaging oxygen  radicals  induced by sustained
 exposure  to this chemical.  DEHP did not appear to directly produce
 repairable DNA damage  in rat  hepatocytes.  The  DNA damage,  as assessed by
 the autoradiographic  measurement  of unscheduled DNA synthesis,  was  not
 observed  in primary rat hepatocytes  exposed  in vitro to DEHP or in  vivo
 by a single  gavage  dose of  5  ^g/kg  DEHP administered 2,  15,  or 24 h
 prior to  the isolation of hepatocytes.

      Sustained feeding of DEHP at a  dietary  concentration  of 20,000 ppm
 led to  a  marked  proliferation of peroxisomes in the liver  after 4 weeks
 of treatment.  Additional administration of a single gavage  dose of  5
 g/kg DEHP to animals  fed the  20,000-ppm diet for  4  or  8  weeks,  as well
 as to 4-week-fed animals that were  also pretreated  with  3-amino-l,2,4-
 triazole  (to inhibit  endogenous catalase activity),  did  not induce  any
 detectable. DNA repair  in hepatocytes  isolated  15  h  following the single
 gavage  dose  of DEHP. Lipid peroxidation in livers of animals treated
 either  with  a  single dose of  5 g/kg  DEHP or with  a  20,000  ppm DEHP  diet
 for  6 weeks, plus a single dose of  5 g/kg, did  not  differ  from controls
 These findings suggest  that DEHP does not  elicit  DNA damage or llpid
 peroxidation in  liver consequent to  the proliferation  of peroxisomes
 resulting from 4  to 8 weeks of administration.

     Lak*-et al.  (1984) observed a marked  species difference  in response
 to peroxisome proliferators. Male Sprague-Dawley  rats  and  Syrian
hamsters were  treated with 25, 100,  250. and 1,000  mg/kg/day DEHP orally
 for  14 days. Liver  enlargement was observed in both  species,  with more
marked effects being observed in  the rat.   Liver weights were  increased
 significantly at doses of 100 mg/kg and greater in  the rat,  but only  at
 1,000 ag/kg  in the hamster. In the rat  there was a marked dose-dependent
 induction of the peroxisomal marker cyanide-insensitive palmitoyl-CoA
oxidation and also of carnitine acetyltransferase. Little effect was
observed on  the mitochondria! markers carnitine palmitoyltransferase  and

-------
76   Section 4

succinace dehydrogenase.  Increased peroxisomal  enzyme activities were
observed after treatment with 100 and 250 mg/kg/day DEHP.

     In contrast, much Less effect was observed in the hamster,  even
after 1,000 mg/kg/day DEHP. Parallel morphological investigations
demonstrated a greater increase in hepatic peroxisome numbers in the rat
than in the hamster. ^C-labeled DEHP was found to be more rapidly
hydrolyzed by rat than hamster hepatic and small intestinal mucosal cell
preparations, and differences were also observed in the absorption and
excretion of oral doses of 14C-DEHP. Studies with MEHP and a
hypolipidemic drug, clofibrate, also resulted in a greater increase in
hepatic peroxisomal enzymes in the rat compared to the hamster.

4.3.6.5  General discussion

     Based on the weight of evidence, there is  sufficient reason to
conclude that DEHP is a carcinogen in experimental animals. DEHP,
administered in the diet, was carcinogenic in rats and mice, producing
increased incidence of liver cancers in female  rats and in male and
female mice. Because DEHP is not mutagenic and  has been demonstrated to
be a peroxisome proliferator, the excess liver  cancers may be due to
peroxisome proliferation. It is unclear whether this effect is likely Co
occur in man, because of differences in'metabolism and liver cell
susceptibility to peroxisome induction, and because structural features
of peroxisomes in humans make these organelles  more difficult to assay.
If peroxisomal induction is the only mechanism of action for tumor
induction in rodents (which is not known), a dose level may exist below
which no tumors would occur. However, current methods of assessing the
carcinogenic potential of chemicals and methods for high-to-low dose and
interspecies extrapolation are not sufficiently sensitive to estimate
such a dose.

4.4  INTERACTION VITH OTHER CHEMICALS
     No studies have been identified which have investigated the effects
of DEHP when administered with other chemicals.

-------
                                                                       77
                5.   MANUFACTURE.  IMPORT.  USE.  AND  DISPOSAL

 5.1  OVERVIEW

      Eighc  major  plants,  locaced mostly  along the  Gulf  Coast  and  in  che
 northeastern states,  produced  approximately  181,700  metric  tons (kkg) of
 DEHP in the United States during 1977. After  accounting for imports  and
 exports,  most of  this total production,  or about  167,600 kkg, was
 available for consumptive use  during  that year. By 1982,  total DEHP
 production  dropped to about 114,000 kkg; in  1986,  production  was back up
 to  131,543  kkg.

      The  major uses of DEHP are  as a  plasticizer  in  polyvinyl chloride
 (PVC)  resins  and  vinyl copolymer resins. Together, these  uses accounted
 for more  than 90%  of  the DEHP  produced in the United States in 1979
 Most of the processing wastes  and by-products containing DEHP are
 disposed  of on land;  relatively  small amounts are  found in wastewater

 5.2  PRODUCTION

      DEHP Is  produced commercially by the esterification of phthalic
 anhydride with  2-ethylhexanol  (octyl  alcohol). The reaction is catalyzed
 by  p-toluene  sulfonic  acid or  by an amphoteric catalyst.  The  acid-
 catalyzed process  involves a water-wash purification step, whereas che
 amphoteric  process does not involve water washing. The  excess alcohol LS
 recovered and  recycled. The product is purified by vacuum distillation
 and/or  with activated  charcoal,  and the yield is >90% (EPA 1981, HSDB
 1987,  IARC  1982).

     There  were eight  major producers of DEHP in the United States in
 1977, operating plants in six  states.  Most of the  total  1977 production
 of  about  181,700 kkg came from two sites (EPA 1981).* Currently, there
 are  eight-major DEHP production  plants (SRI 1987). DEHP  accounted for
 about one-third of the total phchalate ester production  in the United
 States  in 1977. In 1982,  DEHP production was reduced to  114,000 kkg
 (HSDB 1987), but, by 1986, production was 131,543 kkg (EPA 1987b).

 5.3  IMPORT

     No data were located on importation of DEHP into the United States
 in  1977. Approximately 42,500 kkg of phthalate esters were exported  in
 1977. It  is estimated  that about 14,000 kkg of these exports consisted
of DEHP (EPA 1981. HSDB 1987).  The  United States imported 2,722 kkg of
DEHP in 1986 and exported 5,443 kkg (EPA 1987b).
     *This citation erroneously states that Monsanto Company was a 1977
producer of DEHP. Monsanto discontinued DEHP in the early 1970s.

-------
78   Seccion 5

5.4  USE

     The major consumptive use of DEHP is as a plasticizer for PVC
resins (EPA 1981, HSDB 1987). It is also used as a plasticizer for vinyl
chloride copolymer resins. Together, these uses consumed 94% of the DEHP
produced in the United States in 1979 (HSDB 1987). Other reported uses
for DEHP include inert ingredient in pesticides, component of dielectric
fluids (replacing PCBs) in electrical capacitors,  solvent for erasable
ink, acaricide in orchards, vacuum pump oil, and as a testing agent for
air filtration systems (EPA 1981, IARC 1982, HSDB 1987). Of these uses,
the use of DEHP in dielectric fluids is the only one which is.
commercially important and documented.
     Consumer products manufactured from PVC using DEHP as a plasticizer
include vinyl upholstery, tablecloths, shower curtains, raincoats, and
food wrap.  Other products using this material are adhesives, resins and
polymeric coatings, components of paper and paperboard, vinyl gloves for
medical examinations, medical tubing, and flexible bags for intravenous
fluids. Until recently, DEHP was used as a plasticizer in the
manufacture of teething rings, pacifiers, and squeeze toys. Most
manufacturers have discontinued the use of DEHP in these products
(USDHHS 1985).

5.5  DISPOSAL

     Approximately 800 kkg of DEHP were lost to water during the
production processes in 1977. However, most wastes from DEHP processing
and the manufacture of products containing DEHP are disposed of in
landfills.  About 97% of these wastes are disposed of in landfills, and
most of the remaining 3% is incinerated. In 1977,  it is estimated that
materials containing 154,200 kkg of DEHP were disposed of in landfills
(EPA 1981).

-------
                         6.   ENVIRONMENTAL FATE
6.1  OVERVIEW
     DEHP enters the environment as a result of releases during
production and use and after disposal.  With annual U.S.  consumption of
-130 million kg (1986), large quantities of DEHP are available for
potential loss to the environment.  Most DEHP is used as a plascicizer LP
flexible PVC products, typically at levels of 1 to 40% by weight
Applications in a large range of products such as household items,
packaging, and medical products ensures widespread distribution of
plastics containing DEHP.  Losses of DEHP during manufacture are
considered to be around 0.5%. Most  DEHP-containing plastics are disposed
of eventually, with 97% going to landfills and 3% to incineration
Typically, DEHP has been detected in the environment at very low levels

     The environmental fate of DEHP is influenced strongly by its
physical and chemical properties. It is a high-boiling liquid (385°C at
760 nun Hg) with a low water solubility of only 50 to 285 ^g/L and a
vapor pressure of 3.4 x 10"^ nun Hg  (both at 25"C).  The compound is
lipophilic and has a log octanol/water partition coefficient of 4 88
Its biodegradation occurs under aerobic conditions, with a half-life of
several weeks to a month.  Under anaerobic conditions, DEHP degrades much
more slowly. Reaction with hydroxyl radical followed by further
atmospheric chemical reactions, along with deposition and rainout,
removes DEHP from the atmosphere. In the aquatic environment, DEHP
accumulates in sediments,  on suspended solid materials, and in the lip id
tissues of aquatic biota.  In soil,  DEHP is strongly held by soil solids
and organic material such as fulvic acid, from which it is not readily
leached.

6.2  RELEASES TO THE ENVIRONMENT

6.2.1  Anthropogenic Sources
     Table 6.1 summarizes estimates of releases of DEHP to the
environment (EPA 1980) based on the estimated 1986 U.S. production of
130 million kg.

6.2.2  Natural Sources
     Although there have been reports suggesting natural sources of
DEHP, they would be negligible compared to anthropogenic sources  (Mathur
1974. as cited in EPA 1980).

-------
80    Section 6
               Table 6.1. Releases of di(2-ethylhexyl)phlhalate to the environment"

                                                              Amount released*
                              Category                      (millions of kilograms)
U.S. supply
Production waste to water
Transportation loss
Production of products
Vaporized during compounding (loss to air)
Vaporized during compounding (loss to water)
Fabrication waste to landfill
Fabrication waste to incineration
Product inventory
Loss to air
Loss to water
To landfill
To incineration
(130)
0.5
0.1
(129.4)
1.4
2.5
73
0.3
(117.9)
1.8
0.7
112
3.4
                "Based on EPA 1980 adjusted to reflect 1986 U.S. supply of DEHP
           of 130  million kg.
               •*Each figure in parentheses is for a supply/product/inventory
           category: other figures give losses and disposal. For example, of a total
           U.S. supply of 130 million kg, 0.5 million kg is lost as production waste
           to water and O.I  million kg is transportation loss, leaving 129.4  million
           kg for production of products.

-------
                                                 Environmental Face   81

6.3  ENVIRONMENTAL FATE

6.3.1  Atmospheric Fate Processes
     The volatilization of DEHP from plastics and releases during
manufacture and incineration can lead to che presence of chis compound
in the atmosphere  Giam et al.  (1978) reported combined levels of DEHP
of 0 4 ng/ra^ in the troposphere over the Gulf of Mexico and 2 0 ng/m3
over the North Atlantic.  Levels of 300 ng/m3 were reported by Thomas
(1973) near an incinerator in Hamilton,  Ontario, Canada. Because of LCS
adsorption properties (log p - 4.88), atmospheric DEHP should have a
strong tendency to adhere to atmospheric particulate matter,  especially
organic matter and soot,  and to be removed from the atmosphere by
rainout. Chemical hydrolysis of DEHP might occur to a very limited
extent. Direct photolytic reactions are very unlikely. As with most
organic compounds, reaction with the hydroxyl radical 'OH can occur,
such as by abstraction of a hydrogen atom:

                    (DEHP)-H + -OH - (DEHP)- + H20 .

The free radical product, (DEHP)-, would undergo further reactions
leading to the eventual removal of DEHP from the atmosphere.

6.3.2  Surface Water/Groundwaeer Fate Processes
     Reports abound in the scientific literature of the occurrence of
DEHP in surface water and to a lesser extent in groundwater.  In the
absence of a direct pollution source, reported levels in water tend to
be of the order of a mlcrogram per liter.
     The removal of DEHP from water occurs primarily through uptake by
suspended matter, sediments, and biota. Its biodegradation occurs under
aerobic conditions, with a half-life in the range of weeks to a month
Under anaerobic conditions, the compound appears to be biodegraded much
more slowly.
     Because of the very low volatility of DEHP, its volatilization to
the atmosphere from water is likely to be negligible. This is true even
under conditions  (such as aeration) conducive to contaminant
evaporation.
     Sediments play a major role  in determining the fate and perhaps
concentrations of DEHP in surface waters. As noted in EPA  (1980),  the
sediment/water distribution coefficient for DEHP is of  the order  of
4 x 103, indicating a very strong tendency of the compound to adhere  to
sediments. Reported values of DEHP concentrations in sediments vary
widely. Gian and Atlas (1980), reporting on values obtained  from
sediments In the Gulf of Mexico,  the Rhine and  Ijssel rivers, and Lake
Superior, cited values ranging from about 3 Mg/kg Co around  100 mg/kg of
sediment. The more heavily laden  sediments likely provide  a  steady
source of DEHP contamination to  the overlying surface water. The  fate of
DEHP will be influenced by several factors. The race of biodegradation
of DEHP in sediments is variable  and uncertain, and chemical processes
(e.g., as hydrolysis) leading to  the destruction of DEHP in  sediments
are probably negligible.  If  those factors favoring biodegradation are
not present, then DEHP may be transported in  the stream sediments.

-------
82   Section 6

6.3.3  Soil Fate Processes

     The phthalate esters, including DEHP,  have a strong affinity for
soil solids, including the organic,  humic fraction of soil.  The
degradation of DEHP in soil has been summarized by Callahan  (1979).  In
soil under anaerobic conditions,  DEHP degrades, but only slowly.  Under
aerobic conditions in soil, DEHP has been shown to degrade with a half-
Life of several days.  DEHP degrades  in soil under aerobic conditions,
but only slowly, if at all, under anaerobic conditions.  As expected,  the
degradation rate increases with increasing temperature.  Studies with
^C-carbonyl-labeled DEHP in aerobic freshwater hydrosoil yielded a
degradation half-life of -14 days. The degradation was found to be
mediated by microorganisms through an initial hydrolysis to  the phthalic
acid monoester and 2-ethylhexyl alcohol by the action of esterase
enzymes. The next step in the biodegradation process was found to be
oxidative decarboxylation of the exposed carboxyl group. An  intermediate
product in the total degradation process is 1,2-dihydroxybenzene.
Optimal conditions for the degradation of DEHP were found to be within  a
range of pH 7 to 9 and at a temperature of -20"C

     Both the mineral and organic fractions of solid soil particles  tend
to bind DEHP. The high octanol/water partition coefficient of this
compound is indicative of strong binding by soil humus,  and  van der
Waals-type bonds with soil minerals  are favored by the aromatic ring and
carbonyl group on the DEHP molecule.

6.3.4  Biotic Fate Processes

     The biotic fate of DEHP has been summarized by Callahan et al.
(1979). Bioaccumulation followed by  metabolization and biodegradation by
numerous organisms constitutes the biotic fate of DEHP.  A relatively
high log octanol/water partition coefficient is indicative of the high
lipid solubility of DEHP. This has been confirmed by direct  observation
of partitioning into the lipids of both plants and animals.

     DEHP is biosorbed by both single- and multicellular organisms.
Because it is degraded by microorganisms and metabolized by
invertebrates, fish, and other animals, the tendency for DEHP to undergo
bioraagnification is lessened. However, very rapid bioaccumulation and
concentration factors, ranging from  several hundred to several thousand
times the concentration of DEHP in water, have been observed for various
aquatic organisms. The concentration factors appear Co be larger for
smaller aquatic invertebrates (such  as Crustacea and midge larvae) than
for fish. Biomagnification of DEHP appears to be much less than that of
organochlorine compounds.

     DEHP is less biodegradable than short (i.e., <6 carbons) carbon-
chain-length phthalate esters, but more so than several other long-
carbon-chain phthalates (i.e., >7 carbons), and much more so than
persistent organic compounds such as the chlorinated hydrocarbon
pesticides and PCBs. Several microorganisms,  including  the saprophytic
bacterium Serratia, Penicillium Lilaanuns, and fnterobaccer aerogenes.
have been shown to degrade DEHP, and Serracia  can use the compound as a
sole source of carbon and energy  Animals  shown to metabolize DEHP
include fish and rats. Rates of metabolism are much less  than  for
short-chain phthalate esters.

-------
                    7.  POTENTIAL FOR HUMAN EXPOSURE
7.1  OVERVIEW
     DEHP has been detected In air,  water,  soil,  and foods, and in blooc
and plasma handled in tubing and containers that  contain DEHP- as a
plasticizer. For the general population,  the most likely route of
exposure is through contaminated food,  which provides an average of
about 0.3 og/day and a maximum of about 2 mg/day  per individual. Because
of its low solubility in water and its  low vapor  pressure,  exposure co
OEHP in either water or air appears  to  be minimal for most individuals
However, at low levels,  DEHP is a ubiquitous contaminant,and this face
combined with the fact that it is an experimental carcinogen in animals
is cause for some concern.  The greatest potential for reducing exposure
of the general population lies in preventing contact of food with high
fat or oil content with containers and  wrappings  containing DEHP
Because of the compound's high lipophilicity,  such foods may tend co
extract it, thereby enabling exposure to humans ingesting the food  The
FDA currently allows the use of DEHP plasticized  containers or wrappings
only for foods that primarily contain water; therefore, properly
packaged foods are unlikely to become contaminated.  The FDA ruling.
however, does not preclude the possible misuse of DEHP plasticized
containers or wrappings by the consumer.

     A high-risk segment of the population consists of individuals
receiving dialysis treatments or large  quantities of blood that has
contacted DEHP-containing tubing or  containers. Among this population
are hemophiliacs and dialysis patients.

     Workers employed at plants that formulate and process plastics are
also subject to exposure from DEHP.  The total pool of such workers
numbers about 600,000 In the United  States, although only a small
percentage of these workers will likely be highly exposed.

7.2  LEVELS MONITORED OR ESTIMATED IN THE ENVIRONMENT

7.2.L  Levels in Air

     Information is limited regarding the levels  of DEHP in air. Because
DEHP's vapor pressure is very low, exposure from  air is expected co be
minimal. Levels of 0.4 ng/m^ were reported in the air over the Gulf of
Mexico and 2.0 ng/m3 over the North  Atlantic by Giam et al. (1978)
Levels of 300 ng/m^ were reported by Thomas (1973) in the atmosphere
near an incinerator in Hamilton. Ontario, Canada. It is generally
accepted that low levels of phthalates  occur in the atmosphere
throughout the United States, with higher levels  near release sources

-------
84   Seccion 7

     The Inhalation Toxicology Research Institute  (ITRI 1983)  conducted
a vaporization study of eight household consumer products for  the
Consumer Product Safety Commission (CPSP)  in 1983.  The products tested
included window curtain, shower curtain,  nursery pad,  tablecloth, floor
tile, a segment of vinyl flooring, and two types of wallpaper.  A very
low rate of vaporization was reported for  each product.

7.2.2  Levels in Water

     In general, concentrations of DEHP in freshwaters lie within a
range of a fraction of a Mg/L to 10 Mg/L,  although occasionally much
higher values have been observed.  Among the levels that have been
reported are 0.1 to 2.19 Mg/L in Japanese  waters,  1 to 85 pg/L in U.S
water basins surrounded by heavy concentrations of industry, and 0.02 to
0.5 Mg/L in three German estuaries. In a surveillance  study of phthalate
esters in surface waters and sediments in  the United States, Michael et
al. (1984) used rigorously controlled sampling techniques, chemical
analysis, and statistical analysis to determine butylbenzylphthalate in
31 U.S. lakes, rivers, and estuaries in the period 1980 to 1982. Their
detection limit was 0.3 ^g/L. Only three sites yielded levels  above the
detection limit, and these ranged from 0.3 to 0.9  Mg/L. Because of its
widespread use and lesser tendency to degrade, it  may  be anticipated
that somewhat higher levels of DEHP could have been found at these
sites. In a study of the determination of  phthalate esters in  samples
from the marine environment, using combined gas chromatography/mass
spectrometry, Waldock (1983) found 0.058 to 0.078  Mg/L of OEHP in an
estuarine water in the United Kingdom. The values  cited above  are well
within the solubility limit of DEHP, now generally accepted to be about
50 Mg/L at 20"C (a value difficult to determine because of the tendency
of the compound to form colloidal suspensions and  yield erroneously high
values of solubility when determined experimentally).  Low aquatic
concentrations of DEHP are consistent with the well known strong
affinity of this compound for suspended particulate matter, biota, and
sediment.

     Hazardous waste site leachate that enters either surface  water or
groundwater supplies must certainly be considered  as a potential source
of DEHP in water. Leachate from such sites often contains significant
quantities of organohalide compounds such as methylene chloride, 1,2-
dichloroethane, and trichloroethene. Much less information is  available
regarding the levels of phthalate esters in such leachate, a situation
due in pare to the greater difficulties associated with analysis of
phthalate esters compared to organohalides. In addition,  the low water
solubility* of DEHP may impede its appearance in certain leachates  A
study by Ghasserai et al. (1984) showed the presence of DEHP in one of  30
leachates at 11 disposal sites at a level of 200 Mg/L. The availability
of a standard method for determining phthalate esters, including DEHP,
in solid wastes and leachates (EPA  1986d)  should give additional impetus
to obtaining more data on DEHP levels in hazardous waste  site leachates

     Because most DEHP-containing plastics are discarded  to landfills
after use, monitoring leachate from municipal landfills for DEHP may
yield useful information regarding  che exposure potential  from  such
sources. Prior to the enactment of  RCRA. ic was possible  that DEHP

-------
                                        PocenCLaL for Human Exposure   =;

 concaminanc wastes not bound in a polymer matrix were disposed of  n
 ordinary landfills.

 7.2.3  Levels In Soil

      A significant fraction of DEHP entering the atmosphere should end
 up in soil as a result of deposition and rainout  Most of che compound
 should remain with the soil because of strong binding by organic soil
 huraus and van der Waals-type bonds with soil minerals that are favored
 by the aromatic ring and carbonyl group on the DEHP molecule
 Furthermore,  the aerobic conditions and abundance of saprophytic
 microorganisms In soil favor degradation in a soil  medium

      Because  of the factors mentioned above,  accurate determination of
 DEHP In soil  is very difficult,  and this is reflected by a dearth of
 data on the subject.  Despite the large amounts of DEHP deposited in
 landfills,  little information is available on levels of this chemical
 (or other phthalate esters)  in  soils  near landfill  sites  Soil analyses
 have  been reported for samples  taken  In the vicinity of a DEHP factory
 and near  an industrial dump by  Persson et al.  (1978).  Levels of 0 to 0 5
 mg/kg DEHP  in  soil were reported,  although these values  were not
 corrected for  the reported  analytical  recovery (60%  of known analyte
 concentration  in quality  control  samples  was  recovered).

 7.2.4  Levels  in Food

      A  potential exists for  DEHP  contamination of food  during
 processing, handling,  transportation,  and packaging.  This  potential  is
 especially  high  for DEHP-plastlclzed wrappings  on fatty  foods   As  a
 consequence, by  1980.  FDA had allowed  such  wrappings  only  for  foods  •-!-_-
 a high  water content  containing minimal  fat or  oil  (EPA  1980).  In
 addition  to packaging materials, a potential  source  of DEHP  in food  is
 plastic that contacts  the food during  some  stage  of  processing,  for
 example,  in PVC  milk  tubing.

      A  1974 FDA  survey, which included margarine, cheese,  meat,  cereal.
 eggs, milk, white bread, canned corn, corn meal,  and baked beans,  showed
 DEHP  levels in most foods of less than 1  mg/kg.

      In a recent migration study sponsored by  the CMA  (1986),  it was
 shown that  the levels  of DEHP found in fatty  foods such  as milk, cheese
 and meat were not significantly different from background  levels.

 7.2.5  Resulting Exposure Levels

      DEHP is the phthalate to which humans are most likely to  be
 exposed. Exposure of the general population to DEHP Is low, with an
 average of about 0.3 mg/day and a maximum of about 2 rag/day  through
 ingestion of phthalate-contarainated food. For the general population.
 relatively small amounts of DEHP are taken in from drinking water, che
 inhalation of contaminated air.  and skin absorption (EPA 1980)  Higher
exposures occur to people receiving blood handled in plastics,  such as
hemophiliacs and dialysis patients.

-------
86   Section 7

7.3  OCCUPATIONAL EXPOSURES
     A high potential exists for the release  of DEHP  to  the workplace
environment from manufacturing operations.  This potential  is apparent
from an examination of the four basic methods of compounding DEHP
plasticizer with resins.  The methods include  (1) hot  compounding; (2)
dry blending; (3) formation of a stable liquid dispersion  of resin in
plasticizer, known as a plasticol.  and (4)  dissolution of  the resin in a
solvent followed by mixing with a plasticizer to make solvent-cast film
and surface-coating materials. As noted in  Sect. 6. Table  6.1,  about 1 4
million kilograms of DEHP are vaporized to  the atmosphere  during
plastics manufacturing processes (compounding) each year.  An additional
1 8 million kilograms are lost to air from  plastic product inventory
each year.
     Occupational exposures to DEHP may be  relatively high (Liss et al.
1985). Approximately 600,000 U.S. workers are exposed to DEHP in its
manufacture, in plastics formulation, and in manufacturing p.rocesses
that use plastics. In a study of workers in a DEHP formulation plant
(Liss et al. 1985), 6 of 50 personal air monitoring  samples showed
levels of DEHP above the analytical limit of detection (10 ng/sample).
These air samples corresponded to time-weighted average concentrations
ranging from 20 to 4,110 /ig/m3, with a mean of 71 /ig/»3- The urine of
workers was also monitored for total phthalate, showing detectable
levels in the more heavily exposed workers  and significant increases in
samples taken after each shift as compared  to pre-shift samples. It
should be noted that the DEHP levels observed by Liss et al. (1985) were
wichin the  OSHA Permissible Exposure Level  (PEL) of 5 mg/m3.
     Acute  toxic responses are unlikely in workers exposed to DEHP. A
publication (Gosselin 1984) on the clinical toxicology of commercial
products assigns to this compound a toxicity  rating of I — practically
nontoxic. However, the compound has been shown  to cause liver tumors  in
Fischer 344 rats and B6C3F1 mice (Menzer and Nelson 1986, NTP 1982);
therefore,  it is considered to be an experimental animal carcinogen.

7.4  POPULATIONS AT HIGH RISK

7.4.1  Above-Average Exposure
     Exposure of the general population to DEHP  tends to be  low and is
mainly through food sources. Despite low exposures,  the ubiquitous
presence of residues of  this  substance and the  fact  that  it  is  an
experimental animal carcinogen are  reasons for  some  concern.

     Some segments of the  population,  such as  dialysis  patients and
hemophiliacs receiving large  quantities of blood, receive  appreciably
higher levels of DEHP than does  the  general  population, if their
treatment involves the use of  DEHP-plasticized plastics.
     The  largest segment  of  the  population at risk of DEHP exposure
consists of approximately  600.000 workers  engaged in activities such  as
plastics  formulation. The  low  volatility of  DEHP is  helpful in
controlling worker exposure,  especially  through inhalation.

-------
                                       Potential for Human Exposure   *

7.4.2  Above-Avarage Sensitivity

     No Information is available on populations with above-average
sensitivity to DEHP. However,  results from animal studies indicate chat
an age-dependent susceptibility to testicular and hepatic effects ma/
exist,  with immature animals being more susceptible.

-------
                          8.  ANALYTICAL METHODS

      Gas chromatography (GC) is Che mosc common analytical method for
 detecting and measuring DEHP in environmental and biological samples
 High performance liquid chromatography (HPLC) may also be used Co
 measure DEHP in biological samples (Pollack et al.  1985a)

      Samples may be prepared by several methods,  depending on the macr'
 being sampled.  The sample preparation procedures  for analyzing
 environmental samples for DEHP include extraction with carbon dlsulf-de
 extraction with methylene chloride,  soxhlet extraction,  and sonicac'on
 extraction (EPA 1983.  1986c;  NIOSH 1977).  Sample  preparation for
 biological samples often involves  extraction with hexane,  ethyl  acecace
 or chloroform/methanol (Sjoberg et al.  1985a.  Pollack et al  1985a
 Jaeger and Rubin 1972.  Hillman et  al.  1975,  Overturf et  al.  1979)

      Detectors  used to identify DEHP  include the  flame ionization
 detector (FID)  and the electron capture detector  (ECD) (EPA 1983,  1986c
 NIOSH 1977).  When unequivocal  identification is required,  a mass
 spectrometer  (MS)  coupled  to  the GC column  (GC/MS)  may be  used
 (EPA 1983).

      Contamination of  laboratory apparatus  and solvents  with DEHP  is
 very common,  since  it  is a  component of many plastic  and rubber  procuc-s
 and  is  ubiquitous  in  the environment.  Florisil and  alumina column
 clean-up procedures are  recommended to  help  eliminate  interferences, arc
 plastics should  not be used in  phthalate ester analyses  (EPA 1983)

 8.1   ENVIRONMENTAL MEDIA

      Representacive methods appropriate  for  measuring  DEHP in
 environmental media are listed  in Table 8.1.

 8.1.1  Air

     The NIOSH method for measurement of DEHP  in air  involves collector
 of vapors in the air onto a cellulose membrane filter, extraction wi-n
 carbon disulfide. and injection of an aliquot of the extract  into a CC
 equipped with an FID.  The resulting peaks are measured and compared .it-
 standards (NIOSH 1977. as cited  in IARC 1982).

 8.1.2  Water

     The EPA describes two methods (606. 625) for analyzing wastewacer
samples for DEHP. Both of these methods employ packed column GC  for
separation of the organic pollutants,  however, the methods differ in
sample preparation procedures and detection instrumentation.

-------
90
Section  8
                            Tabte«.l.  Analytical
                                               for

Sample
matrix
Air
Sample preparation
Collection on cellulose
membrane niter.
extraction with carbon
disulfide
Analytical
method"
GC/FI
Sample
detection
limit
1 27-6 82 pom
Accuracy
NAe
Reference*
NIOSH 1977.
cited in (ARC
1982
    Water    Extraction with methylene
             chlonde: exchange to
             hexane

             Extraction with methylene
             chloride

    Soil      Soxhlet or somcation
             extraction: exchange to
             hexane
    Food     Extraction with
             acetonitnle and petroleum
             ether
             Extraction with
             acetonitnle. methylene
             chlonde. and petroleum
             ether

             Extraction with hexane.
             acetonitnle, and
             petroleum ether
EPA method 606-    2.0 Mg/L
GC/EC


EPA method 625-    2 5 Mg/L
GC/MS
EPA method 8060   20 Mg/L to
GC/MS or GC/FI   2 g/L*

GC/EC            0.1 ng
                                                                  85%
                                                                                        EPA 1983
                                                                  82%           EPA 1983


                                                                  O.S3C+2.02*    EPA I986c
                               GC/EC
                               GC/FI
 ppb
ISppb
                                                                  90.7%
70-100%
                                   65-70%
                              Thuren 1986
                                                  Giametal  197$
                                                                                 Williamt 1973
        "GC. gai chromatography; Fl. name lonuatnn; EC. electron capture; MS, man ipectromctry.
        * Average percent recovery, unlest otherwue indicated.
        ' Not available.
        rf Depending on the matrix.
        'Here, C - concentration in Mg/L, in an equation directly relating accuracy to concentration.

-------
                                                  Analytical Mechods   9

      Measuring  method  606  is optimized  for  phthalate  esters with a
 detection  limit of  2.0 pg/L for DEHP  Method 625. which employs MS for
 detection  and quantification,  is more broadly applicable  to a large
 number of  base/neutral and acid extractable organics  Sample detection
 limits and accuracy  for DEHP are similar for both methods

 8.1.3  Soil

      Soil  samples can  be analyzed for DEHP by GC with an  ECD or FID
 Sample preparation  is  by soxhlet or sonication extraction, with an
 exchange to hexane. This method (8060)  is used by the EPA for analyzing
 solid waste for phthalate esters.  The method detection limit for DEHP
 ranges from 20  to 2 x  106 ng/L, depending on the matrix (EPA 1986c)

 8.1.4  Food

     The most common method for analyzing foods for DEHP  is by GC/ECD
 (Thuren 1986. Giam et al.  1975).  Analysis may also be by GC/FID
 (Williams  1973,  Ishida et al.  1981)  or gel filtration with GC or HPLC
 (Baker 1978).

 8.2  BIOMEDICAL SAMPLES

     Methods for the analysis of DEHP in biological samples are listed
 in Table 8.2.

 8.2.1  Fluids and Exudates

     Blood levels of DEHP are measured using HPLC with ultraviolet
 absorbance monitoring at 254 run.  The samples are prepared by extraction
with ethyl acetate (Pollack et al.  1985a).  Plasma levels of DEHP may be
measured with GC/MS after extraction with hexane (Sjoberg et al. 19S5a)

     Analysis of urine for DEHP is  by GC/MS (Liss et al.  1985)

8.2.2  Tissues

     Human tissues,  including lung,  liver,  spleen, kidney, heart,  and
adipose,  have been analyzed for DEHP by GC methods (Jaeger and Rubin
 1972, Hillman et al. 1975,  Overturf  ec al.  1979).

-------
92     Section  8
                   Table 8.2. Analytical
for dM2-ethylh*xyl)pfctBaltt* !• Matofkal
Sample
matrix
Unne
Plasma
Whole blood

Lung, liver.
spleen, adipose
Heart

Kidney

Sample preparation
NR*
Extraction with hexane
Extraction with ethyl
acetate
Extraction with
chloroform / methanol
Extraction with
chloroform/methanol
Extraction with
chloroform/methanol
Analytical
method0
GC/MS
GC/MS
HPLC/UV

GC

GC/MS

GC/MS

Sample
detection
limit
NR nmol/mL
3 8 Mg/mL
0 345 Mg/mL

5 0 Mg/gf
dry weight
002Mg/g

0 1 ng/mgc

Accuracy
NR
NR
NR

60-90%

NR

NR

Rcferenca
List et aL 1985
Sjoberg et aL I985»
Pollack et al. 198Sa

Jaeger and Rubin 1972
Rubin 1972
Hillmuetal I97S

Overturf et al. 1979

        "GC. gas chromatography: MS. mass spectrometry. HPLC. high-performance liquid chromatography. UV,
    ultraviolet spectroscopy
        *NR - not reported.
        r Lowest concentration reported.

-------
                    9.   REGULATORY AND ADVISORY STATUS

 9.1  INTERNATIONAL

      The World Health  Organization (WHO)  has not recommended a drin.ki.rg
 water guideline value  for DEHP

 9.2  NATIONAL

 9.2.1  Regulations

      Regulations applicable  to DEHP  address  occupational exposure
 concentrations,  spill  quantities,  presence  in hazardous wastes  and
 reporting rules  (Table 9.1).

      OSHA sets  Permissible Exposure  Limits  (PELs)  for occupational
 exposures to  chemicals based  on the  recommendations  of the  National
 Institute for Occupational Safety  and  Health (NIOSH).  The OSHA PEL  for
 DEHP  is  5 mg/m-}  in workplace  air  for a time-weighted average (TWA)  (3
 h/day. 40 h/week).

      CERCLA requires that persons  in charge  of facilities from which a
 hazardous substance has been  released  in quantities  equal to or greater
 than  its  reportable quantity  (RQ)  immediately notify  the National
 Response  Center  of the  release. The statutory RQ for  DEHP set by che E?-
 Office of Emergency and Remedial Response (OERR) is  1  Ib. However  the
 EPA has proposed revising the  RQ to 100 Ib,  based  on  the assignment of
 DEHP  to Category B in  the hazard-ranking, weight-of-evidence groups for
 potential  carcinogens.

      Chemicals are included on  the RCRA Appendix VIII  list  of hazardous
 constituents  (40 CFR Part 261)  if  they have  toxic,  carcinogenic,
 mutagenic, or teratogenic effects on humans  or other  life forms.  DEHP is
 included  on this list.  Vastes containing DEHP  are  subject to the  RCRA
 regulations promulgated by the  EPA Office of  Solid Waste  (OSW).

     The Office  of Toxic Substances (OTS) promulgates  regulations
 related to manufacturers and/or processors of chemicals  which may
 present an unreasonable risk to health or the environment.  Manufacturers
 of DEHP are required to submit  to EPA information on  the  quantity of :he
 chemical manufactured or imported, the amount directed  to certain uses.
 and the potential exposure and environmental  release of  the  chemical
under the Preliminary Assessment Information Rule.  Unpublished health
and safety studies on DEHP must be submitted to EPA by manufacturers or
processors under the  Health and Safety Data Reporting Rule.

-------
Seccion  9
                        Tabfc9.l. Regolaitoas aad gvidetiM applicable
                                to tfM2-etkylk«iyl)Dejihalau
          Agency
                                Description
                                                          Value
                                                                      References
       OSHA


       EPA OERR
       EPAOSW
       EPAOTS
              Nadosal regulation*

Permissible exposure limit (PEL)
Time-weighted average (TWA)       5 mg/m

Reponable quantity (RQ)
  Statutory                       I l»
  Proposed                         100 Ib

Luting as a Hazardous Waste         NAa
Constituent (Appendix VIII)

Preliminary Assessment Information    NA
Rule

Health and Safety Data Reporting     NA
Rule
29 CFR  1910
(1971)


40 CFR  302.4
50 FR 13456
(04/04/85)
52 FR 8140
(03/16/87)

40 CFR  261
45 FR 33084
(05/19/80)

40 CFR  712
47 FR 26992
(06/22/82)

40 CFR 716
47 FR 38780
(09/02/82)
       NIOSH        Recommended exposure limit (RED   Lowest        NIOSH 1985
                      for occupational exposure            feasible
                                                       limit

       ACGIH        Threshold limit value (TLV)                       ACGIH 1986
                        TWA                          3 mg/m*
                        Short-term exposure limit (STED   10 mg/m1
       NAS          Chronic suggested no adverse         4.2 mg/L      NAS 1977
                      response level (SNARL)
       EPA OWRS    Ambient water quality cntena to                    45 FR 79318
                      protect human health                              (11/28/80)
                        Ingesting water and organisms      15 mg/L
                        Ingesting organisms only           50 mg/L
       EPA OWRS    Draft water quality cntena                         EPA I987e
                        1-day HA for 10-kg child'         20 mg/L
                        1-day HA for 70-kg adult         70 mg/L
                        10-day HA for  10-kg child         S mg/L
                        10-day HA for  70-kg adult         17 S mg/L
                        Lifetime DWEL for 70-kg adult'    I 33 mg/day

       IARC         Carcinogenic rank                 Group 2B     IARC 1982
       EPA          Carcinogenic rank                 Group B2     EPA I986b

State
environmental
agencies
Scat* rtfmtadoei
Water quality standards for
some states

s
3 mg/L Environment
Reporter

           "NA - not available
           0
            HA - health advisory
           f DWEL - drinking water equivalent level

-------
                                      Regulatory and Advisory Scacus   '•:

 922  Advisory Guidance

      Advisory guidance levels are environmental concentrations
 recommended by either regulatory agencies or other organizations which
 are protective of human health or aquatic life  Although not
 enforceable, these levels may be used as the basis for enforceable
 standards. Advisory guidance for DEHP is summarized in Table 9 1 and
 includes the following  a NIOSH Recommended Exposure Limit (REL) for
 occupational exposure, a threshold limit value (TLV).  ambient water
 quality criteria,  a chronic Suggested No Adverse Response Level (SVARL.
 calculated by the  National Academy of Sciences (NAS);  and carclnoaen-c
 ranking by EPA and IARC

      The NIOSH REL for occupational exposure to DEHP in air  is the
 lowest feasible limit. The NIOSH does noc specify an Immediately
 Dangerous to Life  or  Health (IDLH)  value,  but recommends that DEHP be
 treated as a potential human carcinogen.

      The American  Conference of Governmental Industrial Hygienists
 (ACGIH)  recommends a  TLV  TWA of 5  mg/m3  and  a Short-Term Exposure  Limic
 (STEL)  of 10 mg/ra-5 per 15 rain per  8  h based  on the  toxicity  of DEHP

      The  ambient water quality criteria  are  guidelines  set by the  EPA
 Office  of Water Regulations  and Standards  (OWRS)  to  protect  human  healch
 from  potential  adverse effects  from  the  ingestion of water and/or
 organisms,  or  from ingestion of organisms  only from  surface  water
 sources.  The values for DEHP are  15  mg/L  for ingesting  water  and
 organisms  and  50 mg/L for ingesting  only organisms.

     The  NAS calculated a chronic SNARL  for  DEHP  of  4 2  mg/L  based on
 chronic  toxicity data. The calculation was based  on  an  Acceptable  Dail
 Intake  (ADI) of 0  6 mg/kg/day,  assuming 20%  of total intake  is  from
 water, as  follows:

                0.6 mg/kg/day x  70 kg x 0.2
                	  4.2 mg/L  .
                           2  L/day

 9.2.3  Data Analysis

 9.2.3.1  Reference dose

     A reference dose  (RfD) has been calculated for DEHP  (EPA  1987)  It
was based on a 1-year study  in guinea pigs by Carpenter et al.  (1953)
 In this study, groups of 23  to 24 animals of each sex were fed diets
containing 0, 400,  or 1,300 ppm DEHP. This is equivalent  to doses  of -0
 19. and 64 mg/kg/day.  Statistically significant Increases in  relative
 liver weights were  observed  in both groups of  treated animals. An  RfD
was determined as follows:

                    (19 mg/kg/day)
                    	 2 x ID'2 mg/kg/day  .
                        1.000

-------
96   Seccion 9

where

     19 mg/kg/day - NOAEL
     1,000 - uncertainty factor

9 2.3.2  Carcinogenic potency
     Data from the NTP (1982) bioassay (Sect.  4.3  6)  were used by EPA
(1987c) to calculate the upper-bound incremental unit carcinogenic risk
to humans. Using the linearized multistage  model,  potency slope factors
were estimated.  These are shown below.

            Animals on which based
            	        Slope factor3
            Species          Sex            [(mg/kg/day)'1]
Rat
Mouse
Male
Female
Male
Female
0.0045
0.0035
0.0084
0.01
              ^Calculations were based on combined carcinomas
            and neoplastic nodules (or adenomas).

     The unit risk value is estimated to be 4.0 x 10"7 for drinking
water containing 1 Atg/L DEHP  The drinking water concentrations of DEH?
corresponding to lifetime excess cancer risks of 10'4, 10'5, and 10'6
are 0.3, 0 03, and 0.003 mg/L, respectively.
     In the NTP (1982) bioassay, an adequate number of animals were
observed,  and a statistically significant increase in incidence of liver
tumors was seen in both sexes and was dose dependent in both sexes of
mice and in female rats.
     The CPSC (1985) estimated the lifetime excess risk per mg/kg/day
for hepatocellular carcinomas and neoplastic nodules observed in the NTP
(1982) study. The estimates of risk to humans using the linearized
multistage model with body-surface-area correction factors of 5.6 for
rats and 12.8 for mice are as follows:

                     Lifetime human risk estimates
                            [(mg/kg/day)"I]
Animal on which based
Species
Rat
Mouse
Sex
Male
Female
Male
Female
Maximum
likelihood
estimate
0 000862
0 00160
0 00868
0 00698
Upper 95%
confidence
limit
0 00264
0.00293
0.0144
0.00963

-------
                                     Regulacory and Advisor:-

     The differences between the potency slope factors calculated b/ £?-
(1987c) and CPSC's (1985) upper 95% confidence limit values are due :o
differences in doses used in the two agencies' calculations

9.3  STATE

     Regulations and advisory guidance from the states were still be:-g
compiled at the time of printing

9.3.1  Regulations

     Phthalate esters are specified in a few State Water Quality
Standards; the standard for public water systems is 3 Mg/L. .'-."hen not
specified, DEHP is included in the category "toxic substances" in
narrative form in the Water Quality Standards of most states.  Specific
narrative standards protect the use of surface waters for public -acer
supply and contact recreation.

9.3.2  Advisory Guidance

     No data were available on state advisory guidance

-------
                            10.   REFERENCES


ACGIH (American Conference of Government Industrial Hygienists).  1986
Documentation of the Threshold Limit Values and Biological Exposure
Indices. 5th ed. Cincinnati,  OH.

Agarwal DK, Eustis S. Lamb JC. Reel JR.  Kluwe WM  1986.  Effects of di-
(2-echylhexyDphchalace on the gonadal pathophysiology,  sperm
morphology, and reproductive  performance of male rats.  Environ Healch
Perspect 65:343-350.

Agarval DK, Agarwal S,  Seth PK.  1982.  Effect of DEHP on drug metabolism.
lipid peroxidation. and sulfhydryl content of rat liver.  Drug Metab
Dispos 10:77-80.

* Albro PW. 1986. Absorption, metabolism, and excretion of DEHP by ra-s
and mice. Environ Health Perspect 65:293-298.

Albro PV, Corbett JT, Schroeder JL, Jordan S, Matthews HB. 1982.
Pharmacokinetics, interactions with macromolecules, and species
differences in metabolism of  DEHP. Environ Health Perspect 45:19-25

Albro PW, Tondeur I, Marbury  D,  Jordan S, Schroeder J, Corbect JT.
1983a.  Polar metabolites of  DEHP in the rat. Biochim. Biophys. Acca
760: 283-292.

Albro PW, Hass JR, Peck CC, Jordan ST. Corbett JT. Schroeder J. 1983b
Applications of isotope differentiation for metabolic studies with DEK?
J Environ Scl 17:701-714.

Albro PU, Hass JR, Peck CC, et al. 1981. Identification of the
metabolites of DEHP In urine  from the African green monkey. Drug Metab
Dispos 9:223-225.

Albro PV, Thomas R, Fishbein L.  1973.  Metabolism of diethylhexyl
phthalaCe by rats: Isolation and characterization of the  urinary
metabolites. J Chromatogr 76-321-330.

Albro PW, Tondeur I, Marbury D. Jordan S, Schroeder J, Corbett JT. 198^
Polar metabolites of DEHP in che rat.  Biochim Biophys Acta 760:283-292

Aldrich. 1987. Aldrich Chemical Catalog. Aldrich Chemical Company.
*Key studies.

-------
100   Seccion 10

APHA (American Public Health Association)   1985  Standard Methods  for
the Examination of Water and Wastewater  16th  ed  Washington,  DC.

Ashby  1986. The prospects for a simplified and internationally
harmonized approach to the detection of possible human  carcinogens  and
mutagens  Mutagenesis 1:3-16

Ashby J, de Serres FJ,  Draper M. et al  1985.  Overview  and conclusions
of the IPCS collaborative study on in vitro assay systems.  Prog  Hutat
Res 5 117-174

Bailey GL  1977. The sick kidney and sex.  N Eng J Med 296:1288-1289.

Baker RWR. 1978. Gel filtration of phchalate esters.  J  Chromatogr
154 3-11

Barnes D et al. 1987. Reference dose (RfD): Description and use  in
health risk assessments. Appendix A in Integrated Risk  Information
System Support Documentation. Vol. 1. Office of Health  and Environmental
Assessment, Environmental Protection Agency, Washington,  DC.  EPA/600-8-
86/032a.

Bickers DR, Dutta-Choudhury T. Mukhtar H.  1982. Epidermis:  A site  of
drug metabolism in neonatal rat skin. Studies  on cytochrome P-450
content and mixed-function oxidase and epoxide hydrolase activity.  Mol
Pharmacol 21:239-247.

Branson, DR.  1980.  Prioritization of chemicals according to degree of
hazard in the aquatic environment. Environ Health Perspect 34: 133-138

Bratt H, Batten P. 1982. Imperial Chemical Industries PLC (cited in CPSC
1985).

Butterworth BE. 1984. The genetic toxicology of DEHP. Chem Ind Inst
Toxicol (CUT)' 4:1-8.

Callahan MA et al. 1979. Water Related Fate of 129 Priority Pollutants
Environmental Protection Agency, Washington, DC. EPA-440/4-79-029b.

Galley D, Autian J, Guess W. 1966  Toxicology of a series of phthalate
esters. J Pharra Sci 55:158.

* Carpenter CP, Weil CS, Smyth HF. 1953. Chronic oral toxicity of DEHP
for rats, guinea pigs, and dogs  Am Med Assoc  Arch Ind Hyg 8:219-226
(cited in EPA 1987).

CEFIC. 1982  Final reports from Council European of  the Federations of
the Industry Chemical (CEFIC) on Di•2-echylhexyl phthalate (DEHP)

Chadwick M. Branfman AR, Silveira DM   1982  Dose Dependence of and
effect of prior exposure on  the metabolism of  DEHP administered in the
diet to rats  Report to  the Chemical Manufacturers Association. Arthur
D  Little.  Inc  Unpublished study

-------
                                                        References   101

Chadwick M, Branfman AR and Silveira DM. 1983. Species differences in
the metabolism of a single oral dose of DEHP. Report to the Chemical
Manufacturers Association. Arthur D. Little,  Inc.  Unpublished study.

CHRIS (Chemical Hazard Response Information System). 1978.  Data Manual.
U.S. Coast Guard (cited in HSDB 1987).

Clayton CG, Clayton FE. eds.  1981. Patty's Industrial Hygiene and
Toxicology. 3rd revised ed. Vol. IIA. New York.  NY:  John Wiley and Sons.

CMA (Chemical Manufacturers Association).  1982.  Phthalate Esters Program
Panel, Voluntary Test Program,  Health Effects Testing.  Phase I:
Validation Results.  Vol. I. Washington DC,  p. 402.

CMA (Chemical Manufacturers Association).  1983.  Measurement of the water
solubility of phthalate esters. Final report, contract PE-11.  0-WS-SRL.
L1533-06, Syracuse Research Corp., April 1983.

CMA (Chemical Manufacturers Association).  1984.  A 21-Day Dose
Relationship Study of DEHP in Rats.  Report dated July.

CMA (Chemical Manufacturers Association).  1985a.  A 21-Day Feeding Study
of Butyl Benzyl Phthalate in Rats. Effects on the liver and liver
lipids.  Report dated October.

CMA (Chemical Manufacturers Association).  1985b.  A 21-Day Feeding Study
of 610 Phthalate in Rats.  Effects on the liver and liver lipids. Report
dated December.

CMA (Chemical Manufacturers Association).  1985c.  A 21-Day Feeding Study
of 711 Phthalate in Rats.  Effects on the liver and liver lipids. Report
dated November.

CMA (Chemical Manufacturers Association).  1985d.  A 21-Day Feeding Study
of Dlundecyl Phthalate in Rats. Effects on the liver and liver lipids.
Report dated September.

CMA (Chemical Manufacturers Association).  1986a.  A 21-Day Feeding Study
of Di(2-ethylhexyl)adipate in Rats.  Effects on the liver and liver
lipids.  Report dated February.

CMA (Chemical Manufacturers Association).  1986b.  A 21-Day Feeding Study
of Dl-n-butyl Phthalate in Rats. Effects on the liver arid liver lipids.
Report d*ted February.

CMA (Chemical Manufacturers Association).  1986c.  A 21-Day Feeding Study
of Di-isononyl Phthalate in Rats. Effects on the liver and liver lipids.
Report dated January.

CMA (Chemical Manufacturers Association).  1986d.  A 21-Day Feeding Study
of Di-isodecyl Phthalate in Rats. Effects on the liver and liver lipids
Report dated January.

-------
102   Seccion 10

Cohen AJ,  Grasso P.  1981.  Review of the hepacic  response to
hypo I ip identic drugs  in rodents and assessment  of ics  toxicological
significance to Man. Food Cosmet Toxicol 19:585-606.

CPSC (Consumer Product Safety Commission).  1985.  Report to the Consumer
Produce Safety Commission by the Chronic Hazard  Advisory Panel on DEHP

DeAngelo AB. Garrett CT, Manolukas LA,  Yario T.  1986.  Di-n-octyl
phthalate (OOP), a relatively ineffective peroxisome-inducing straight
chain isomer of the environmental contaminant  DEHP,  enhances the
development of putative preneoplastic lesions  in rat  liver. Toxicology
41:279-288.

DeAngelo AB, Garrett CT. Queral AE. Irvin D. 1985a.  Phosphorylation of
specific rat plasma membrane proteins during promotion of gamma-glutamyL
transpeptidase-positive hepatic foci and Inhibition by DEHP. Cancer Res
45:2654-2660.

DeAngelo AB, Queral AE, Garrett CT. 1985b.  Concentration-dependent
inhibition of development of GGT positive foci in rat liver by the
environmental contaminant DEHP. Environ Health Perspect 60:381-385.

De La Iglesia FA, Sturgess JM, Feuer G. 1982.  New approaches for the
assessment of hepatotoxicity by means of quantitative functional-
morphological interrelationships.  In: Plaa G,  Hewitt WR. eds. Toxicology
of the Liver. Raven Press, New York, pp. 47-102.

Diwan BA. Ward JM, Colburn NH, et  al. 1983. Promoting effects of DEHP in
mouse liver, skin, and JB-6 epithelial cells.  Proc Ann Meet Am Assoc
Cancer Res  24:105.

Diwan BA, Ward JM, Rice JM, Colburn NH, Spangler EF. 1985. Tumor-
promoting effects of DEHP in JB6 mouse epidermal cells  and mouse skin.
Carcinogenesis  6:343-397.

*  Dostal LA. Jenkins WL, Schwetz BA. 1987.  Hepatic peroxisome
proliferation and hypolipldemic effects of  DEHP  in neonatal  and  adult
rats. Toxicol Appl  Pharmacol  87:81-90.

Douglas GR,  Hugenholtz  AP,  Blakey  DH.  1986. Genetic toxicology  of
phthalate  esters: Mutagenic and other  genotoxic  effects.  Environ Health
Perspecc 65:255-262.

El Sisi AE.  Carter  DE.  Sipes  1C.  1985.  Dermal absorption and tissue
distribution of phthalate esters.  Toxicologist  5:246.

EPA (Environmental  Protection Agency).  1980.  An Exposure and Risk
Assessment for  Phthalate  Esters.  Environmental  Protection Agency,
Washington,  DC.

EPA (Environmental  Protection Agency)   1981.  An Exposure and Risk
Assessment for Phthalate  Esters.  Final draft  report.  Office of Water
Regulations and Standards,  Washington. DC

-------
                                                        References   101

EPA  (Environmental Protection Agency). L983. Methods for Chemical
Analysis of Water and Wastes. Office of Research and Development,
Environmental Monitoring and Support Laboratory, Cincinnati, OH. EPA-
600/^-79-020.

EPA  (Environmental Protection Agency). 1986a. Guidelines for
carcinogenic risk assessment. Fed Regist 51.33992-34003.

* EPA (Environmental Protection Agency).  1986b. Evaluation of the
Potential Carcinogenicity of Bis(2-ethylhexyl) phthalate. Draft reporc
Carcinogen Assessment Group, Office of Health and Environmental
Assessment. OHEA-C-073-45.

EPA  (Environmental Protection Agency).  1986c. Test Methods for
Evaluating Solid Waste.  3rd ed. Office of Solid Waste and Emergency
Response, Washington, DC. SW-846.

EPA  (Environmental Protection Agency).  1986d. Method 8060, Phthalate
Esters.  Test Methods for Evaluating Solid Waste. 3rd ed. Office of Solid
Waste and Emergency Response, Washington,  DC. SW-846.

EPA  (Environmental Protection Agency).  1986e. Drinking Water Criteria
Document for Phthalic Acid Esters (PA-E)s.  External review draft. ECAO-
CIN-D009.

* EPA (Environmental Protection Agency).  1987a. Integrated Risk
Information System.  Chemical Files. Vol.  II. Office of Health and
Environmental Assessment. EPA/600/8-86/032b.

EPA  (Environmental Protection Agency).  1987b. Dioctylphthalate.  Chemical
Products Synopsis. Office of Toxic Substances.

EPA  (Environmental Protection Agency).  1987c. Integrated Risk
Information System (IRIS).  Risk Estimate  for Carcinogenicity for Di-2-
Ethylhexyl Phthalate (Verification date October 7, 1987). Office of
Health and. Environmental Assessment, Environmental Criteria and
Assessment Office, Cincinnati, OH.

EPA  (Environmental Protection Agency).  1988. Drinking Water Criteria
Document for Phthalic Acid Esters (PAE)s.  Prepared by the Office of
Health and Environmental Assessment. Environmental Criteria and
Assessment Office, Cincinnati, OH, for the Office of Drinking Water,
Washington, DC.  Final draft.

Eriksson P. Darnerud PO. 1985. Distribution and retention of some
chlorinated hydrocarbons and a phthalate  in the mouse brain during the
pre-weaning period.  Toxicology 37:189-203.

Gangolli SD. 1982. Testicular effects of phthalate esters. Environ
Health Perspect 45:77-84.

Canning AE, Brunk U,  Dallnec G. 1984. Phthalate esters and their effecc
on the liver. Hepatology 4:541-547.

-------
104   Section 10

Ghassemi M, Qulnllvan S. Bachmaier J.  1984.  Characteristics of leachat&s
from hazardous waste landfills. J Environ Sci Health 19:579-620.

Giara CS, Adas EL. 1980. Accumulation of phthalate ester plasticizers in
Lake Constance sediments. Naturwissenschaften 67:598.

Ciam CS, Chan HS, Neff CS.  1975. Sensitive method for  determination of
phthalate ester plasticizers in open-ocean biota samples.  Anal Chem
47:2225-2228.

Giam CS, Chan HS, Neff GS,  Atlas EL.  1978.  Phchalate ester plasticizers
A new class of marine pollutant. Science 199:419-421.

Gollamudi R, Rao RH, Lawrence WH, Autlan J.  1985. Effects  of phthalic
acid esters on drug metabolizing enzymes of rat liver.  J Appl Toxicol
5:368-371.

Gollamudi R. Prasanna HR, Rado RH, Lawrence WH, Autian J.  1983.  Impaired
metabolism of DEHP in old rats: An in vitro study. J Toxicol Environ
Health 12:623-632.

Gosselin RE, Smith RP, Hodge HC. 1984.  Clinical toxicology of commercial
produces. Williams and Wilkins, Baltimore/London 11:204.

Gray TJB. Gangolli SO. 1986. Aspects of the testicular toxicity of
phthalate esters. Environ Health Perspect 65:229-235.

* Gray TJ, Buctervorth KR.  1980. Testicular atrophy produced by
phthalate esters. Arch Toxicol (Suppl)  4:452-455.

Gray TJ, Rowland IR, Foster PM, Gangolli SD. 1982. Species differences
in the testicular toxicity of phthalate esters. Toxicol Lett 11:141-147

Harris RS, Hodge HC, Maynard EA, Blanchat HJ. 1956. Chronic oral
toxic icy of 2-ethylhexyL phthalate in rats and dogs. Arch Ind Health
13:259-264.

Haseman JK. 1983. Patterns of tumor incidence in two-year cancer
bioassay feeding studies in Fischer 344 rats. Fundam Appl Toxicol 3:1-9

Haseman JK. 1985. Issues in carcinogenicity testing: Dose selection.
Fundam Appl Toxicol 5:66-78.

Havel RJ, Kane JP. 1973. Drugs and lipid metabolism. Ann Rev Pharmacol
13:287-308.

Hillman LS, Goodwin SL,  Sherman WR. 1975. Identification and measurement
of plasticizer in neonatal tissues after umbilical catheters and blood
products. N Eng J Med 292:381-386.

Hodge H. 1943. Acute toxicity  for rats and mice  of 2-ethylhexanol and
DEHP. Proc Soc Exp Med  53:20-23.

-------
                                                        References

Hopkins J. 1983  Is di-ethylhexyl phchalate genotoxic7 Food Chem TOXLCO.
21:684-687

HSDB (Hazardous Substances Daca Bank)   1987  Record for Bis(2-
echylhexyl)-phthalace Computer Printout  National Library of Medicine
April 9.

IARC (International Agency for Research on Cancer)   1982  Di(2-
ethylhexyl)phthalate  IARC Monogr Eval Carcinogenic Risk Chem Humans
29 269-294.

Ishida M. Suyania K, Adachi S.  1981.  Occurrence of  dibutyl and DEHP IP
chicken eggs.  J Agric Food Chem 29.72-74.

Jacobson MS,  Kevy SV, Grand RJ.  1977.  Effects of a  plasticizer leached
from polyvinyl chloride on the subhuman primate1  A  consequence of
chronic transfusion therapy.  J Lab Clin Med 89 1066-1079.

Jaeger RJ, Rubin RJ  1972. Migration of a phchalate ester plasticizer
from polyvinyl chloride blood bags into stored human blood and its
localization in human tissues.  N Eng J Med 287 1114-1118

* Kluwe WM, Haseman JK. Douglas JF.  Huff JE.  I982a. The carcinogeni.ci.cy
of dietary DEHP in Fischer 344 rats and B6C3F(1) mice  J Toxicol Environ
Health 10:797-815.

* Kluwe WM, McConnell EE,  Huff JE, Haseman JK, Douglas JF. 1982b
Carcinogenicity testing of phthalate esters and related compounds by :r,e
National Toxicology Program and the National Cancer Institute
Conference on Phthalates,  Washington,  DC,  June 9-11, 1982. Environ
Health Perspect 45:129-134.

* Kluwe WM. Haseman JK. Huff JE. 1983. The carcinogenicity of DEHP in
perspective.  J Toxicol Environ Health 12:149-169.

Kornbrusc DJ.  Barfknecht TR,  Ingram P, Shelburne JD. 1984. Effect of
DEHP on DNA repair and lipid peroxidation in rat hepatocytes and on
metabolic cooperation in Chinese hamster V-79 cells. J Toxicol Environ
Health 13:99-116.

Kozumbo WJ, Kroll R, Rubin RJ. 1982. Assessment of the mutagenicity of
phthalate esters. Environ Health Perspect 45:103-109

Krauskopf LG.  1973. Studies on the toxicity of phthalates via ingestion
Environ Health Perspect 3.61-72.

Lake BG, Gray TJ. 1985. Special differences in hepatic peroxisome
proliferation. Biochem Soc Trans 13 859-861.

Lake BG. Gray TJ. Foster JR.  Stubberfield GR. Gangolli SD. 1984.
Comparative studies on DEHP induced hepatic peroxisome proliferation in
the rat and hamster. Toxicol Appl Pharmacol 72:46-60.

-------
106   Seccion 10

Lake BG.  Gray TJ,  Gangolli SD.  L986   Hepatic  effects  of  phthalate  esters
and related compounds - in vivo and  in vitro  correlations.  Environ
Health Perspect 67:283-290.

Lake BG,  Rijcken WRP. Gray TJB. Foster JR,  Gangolli SD.  1983.
Comparative studies of the hepatic effects  of di-  and mono-n-octyl
phthalates, DEHP,  and clofibrate in  the rat  Acta  Pharmacol Toxicol
54 167-176.

Lake BG.  Tredger JM. Gray TJB,  et al.  1984  The  effect of  peroxisome
proliferators on the metabolism and  spectral  interaction of endogenous
substrates of cytochrome P-450  in rat hepatic microsomes  Life  Sci
35-2621-2626.

Lalwani ND. Fahl WE, Reddy JK.  1983.  Detection of  a nafenopin binding
protein in rat liver cytosol associated with  the induction of peroxide
proliferation by hypolipidemic  compounds. Biochem  Biophys  Res Commun
116:388-393.

Lawrence W, Malik M, Turner J,  Singh A, Autian J.  1975.  A  toxicological
investigation of some acute, short-term and chronic effects of
administering DEHP and other phthalate esters. Environ Res 9:1-11

Lazarow PB, DeDuve C. 1976.  A fatty  acyl-CoA  oxidizing system in rat
liver peroxisomes; enhancement  by clofibrate, a  hypolipidemic drug  Proc
Natl Acad Sci 73:2043-2046.

Lhugenot JC.  Mitchell AM, Milner G.  Lock EA,  Elcombe  CR. 1985.  The
metabolism of DEHP and MEHP in  rats:  in vivo  and in vitro  dose  and time
dependency of metabolism. Toxicol Appl Pharmacol 80:11-22.

Lindgren I, Lindquist NG. Lyden A,  Olsson T,  Ullberg  S  1982. A whole
body autoradiographic study on  the  distribution  of ^C-labelled DEHP in
mice. Toxicology 23:149-158.

Liss GM,  Albro PW, Hartle RW, Stringer WT.  1985. Urine phthalate
determinations as an index of occupational  exposure  to phthalic
anhydride and DEHP. Scand J Work Environ Health  11:381-387.

Mangham BA, Foster JR, Lake BG. 1981. Comparison of the  hepatic and
testicular effects of orally administered DEHP and dialkyl 79 phthalace
in the rat. Toxicol Appl Pharmacol  61 205-214.

Mathur S. 1974. Phthalate esters in the environment:  Pollutants or
natural products? J Environ Quality 3 189.

Melnick RL, Schiller CM. 1985.  Effect of phthalate esters on energy
coupling and succinate oxidation in rat liver mitochondria. Toxicology
34:13-27.

Menzer RE, Nelson JO.  1986. Water and soil  pollutants.  In: Klaassen CD.
Amdur MO, Doull J, eds.  Toxicology  3rd ed  New York: Macmillan.  pp
841-842.

-------
                                                        References   107

Michael PR, Adams WJ.  Werner AF,  Hicks 0.  1984. Surveillance of
phchalate esters In surface waters and sediments in the U.S. Environ
Toxicol Chera 3:377.

Mitchell AM, Lhugenot  JC,  Bridges JU,  Elcorae CR. 1985.  Identification of
the proximate peroxisome proliferator(s)  derived from DEHP.  Toxicol Appl
Pharraacol 80-23-32.

* Mitchell FE,  Price SC, Hinton RH,  Grasso P.  Bridges J.W.  1985.  Time
and dose-response study of the effects on rats of the plasticizer DEHP
Toxicol Appl Pharmacol 81:371-392.

Morton SJ.  1979. Hepatic effects  of dietary DEHP.  Ph.D. thesis  Johns
Hopkins University, Baltimore, MD.

Nakamura Y, Yasuoki Y, Toraita I,  Tsuchikawa. 1979.  Teratogenicity of
DEHP in mice. Toxicol  Lett 4:113-117.

NAS (National Academy  of Sciences).  1977.  Drinking Water and Health
Washington, DC:  National Academy  of Sciences,  pp.  726-729.

NFPA (National Fire Protection Association). 1978.  Fire Protection Guide
for Hazardous Materials (cited in HSDB 1987).

NIOSH (National  Institute for Occupational Safety and Health). 1985.
NIOSH Pocket Guide to  Chemical Hazards. Washington, DC: U.S. Department
of Health and Human Services.

NIOSH (National  Institute for Occupational Safety and Health). 1977
NIOSH Manual of  Analytical Methods.  2nd ed. Vol. 2. Cincinnati, OH, pp
S40-1-S40-9.

* Northrup S, Martls L, Ulbrlcht R,  Garber J,  Mlrlpol J. 1982. Comment
on the carcinogenic potential of bis(2-ethylhexyl)phalate.  J Toxicol
Environ Health 10:493-518.

* NTP (National  Toxicology Program). 1982. Carcinogenesis Bioassay of
DEHP (Cas No. 117-81-7) in F344 Rats and B6C3F1 Mice (Feed Study).
National Toxicology Program, Research Triangle Park, NC. NTP-80-37. NIH
Publication 82-1772.

Oishi S, Hiraga  K. 1980. Effect of phthalic esters on mouse  testes.
Toxicol Lett 4:413-416.

Oishi S. 198S.  Reversibility of testicular atrophy induced by DEHP  in
rats. Environ Res 36:160-169.

Oishi S, Hiraga  K. 1982. Distribution and elimination of DEHP and MEHP
after a single oral administration of DEHP  in  rats. Arch Toxicol
51:149-155.

Oishi S, Hiraga  K. 1983. Testicular atrophy induced by  DEHP:  Effect  of
zinc supplement. Toxicol Appl Pharmacol 70:43-48.

-------
108   Seccion 10

Overcurf ML. Drullhet RE. Liehr JG. Kivkendell WM, Captrioli RM. 1979.
Phthalate esters in normal and pathological human kidneys. Bull Environ
Contain Toxicol 22: 536- 54^2.

Parmar D, Srivastava SP, Seth PK  1985  Hepatic mixed function oxidases
and cytochrome P-450 contents in rat pups exposed to DEHP through
mother's milk  Drug Me tab Dispos 13 368-370.

Parmar D, Srivastava SP, Seth PK. 1986. Effect of DEHP on
spermatogenesis in adult rats. Toxicology 42-47-55.

Parry JM, Arni P, Brooks T, Carere A,  Ferguson L, Heinisch J  1985
Summary report on the performance of the yeast and aspergillus assays
Prog Mutat Res 5:25-46.

Patty F. 1967. Industrial Hygiene and Toxicology. Vol. II. New York:
Interscience Publishers, pp. 1904-1906.

Peck CC, Albro PW. 1982. Toxic potential of the plasticizer DEHP in the
context of its disposition and metabolism in primates and man. Environ
Health Perspect 45:11-17.

Pegg DG  1979. Toxicity and biologic fate of DEHP following inhalation
exposure in rats  Research Report 81-135. Unpublished study. August 8.

Persson P, Penttinen H, Nvortera P. 1978. DEHP in the vicinity of an
industrial area in Finland  Environ Pollut 16:163.

Peterson A, Draman A, All P, Vinyard D  1974.  Toxicity of Plastic
Devices Having Contact with Blood. National Technical Information
Service. U.S. Department of Commerce.  Washington, DC. NTIS Publication
P3-233701.

Phillips BJ, James TTB, Gangolli SD. 1982. Genotoxicity studies of DEHP
and its metabolites in CHO cells. Mutat Res 102:297-304.

Pollack CM, Buchannan JF. Slaughter RL, Kohli RK. Shen DD. 1985a.
Circulating concentrations of DEHP and its de-esterified phthalic acid
products following plasticizer exposure in patients receiving
hemodialysis. Toxicol Appl Pharmacol 79 257-267

Pollack GM. Li RC, Ermer JC, Shen DD.  1985b.  Effects of route of
administration and repetitive dosing on the disposition kinetics of DEHP
and its mono-de-esterified metabolite in rats. Toxicol Appl Pharmacol
79.246-256.

Price CJ, Tyl RW, Marr MC, Sadler BM,  Kimmel CA  1986. Reproduction and
Fertility Evaluation of Diethylhexyl phchalate (CAS No. 117-81-7) in
Fischer 344 rats Exposed During Gestation  Final report. National
Toxicology Program. National Institute of Environmental Health Sciences
NTP-86-309.

-------
                                                        References   L09

Prlscon FAJ,  Dean BJ.  1985.  Tests for che  induction of chromosome
aberrations,  polyploidy,  and sister chromatid exchanges in rat Liver
(RL4) cells.  Prog Mutat Res  5.387-395.

Probst CS.  Hill LE.  1985.  Tests  for the  induction of DNA repair
synthesis in primary cultures of adult  rat hepatocytes. Prog Mutat Res
5.381-386.

Rao MS. Reddy JK. 1987. Peroxisome proliferation and
hepatocarcinogenesis.  Carcinogenesis 8:637-645.

Reddy JK. 1987. Cell specificity in the  induction of peroxisome
proliferation. In: Dahlem Workshop on Mechanisms of Cell Injury (in
press).

Reddy JK, Laiwani ND.  1983.  Carcinogenesis by hepatic peroxisome
proliferators: Evaluation of the risk of hypolipidemic drugs and
industrial plasticizers to humans. Crit  Rev Toxicol 12:1-58.

Reddy JK, Moody DE,  Azarnoff DL, Rao JK. 1976. Di(2-ethylhexyl)-
phthalate:  An industrial plasticizer induces hypolipidemia and enhances
hepatic catalase and carnitine acetyltransferase activities in rats and
mice. Life Sci 18:941-946.

Reddy JK, Reddy HK.  Usman MI, Lalwani NO,  Rao MS. 1986. Comparison of
hepatic peroxisome proliferative effect and its implication for
hepatocarcinogenicity of phthalate esters, DEHP and di(2-ethylhexyl)-
adipate, with a hypolipidemic drug. Environ Health Perspect 65:317-327

Reddy JK, Reddy MK,  Usman MI, Lalwani ND,  Rao MS. 1986. Comparison of
hepatic peroxisome proliferative effect and its implication for
hepatocarcinogenicity of phthalate esters, DEHP and di(2-ethylhexyl)-
adipate, with a hypolipidemic drug. Environ Health Perspect 65:299-307

* Reel JR, Lawton AD, Lamb JC 1982. Diethylhexyl phthalate  (DEHP):
Reproduction and Fertility Assessment in CD-I Mice When Administered in
the Feed. Report ISS RTI-31U-2344. RTI-72, NTP-84-079. Order  PB84-
181734, p. 221.

* Rhodes C, Orton TC, Pratt IS.  Batten PL, Bratc H. Jackson SJ.  1986
Comparative pharmacokinetics and  subacute toxicity of  DEHP  in racs and
marmosets: Extrapolation of effects in rodents  to man. Environ Health
Perspect 65:299-308.

Sanner T, Rivedal E. 1985. Tests  with the Syrian hamster  embryo  (SHE)
cell transformation assay. Prog Mutat Res 5:665-671.

Saxena DK, Srivastava SP, Chandra SV, Seth PK.  1985. Testicular  effects
of DEHP: Histochemical and histopathological  alterations.  Ind Health
23:191-198.

Schmid P, Schlatter C. 1985.  Excretion  and metabolism  of  DEHP in man
Xenobiotica  15:251-256.

-------
110   Section 10

Schulz CO,  Rubin RJ.  1973.  Distribution, metabolism,  and  excretion  of
DEHP in the rat. Environ Health Perspect 3:123-129.

* Shaffer CB, Carpenter CP,  Smyth  HF.  1945. Acute  and subacute  toxicity
of DEHP with note upon its  metabolism. J Ind Hyg Toxicol  27:130-135.

Shiota K, Mima S. 1985. Assessment of  the  teratogenicity  of DEHP  and
MEHP in mice. Arch Toxicol  56:263-266.

Shiota K, Chou HJ,  Nlshimura H. 1980.  Embryotoxic  effects of DEHP and
di-n-butyl phthalate (DB) in mice.  Environ Res  22:245-253.

Singh AR, Lawrence WH, Autlan J. 1972. Teratogenicity of  phthalate
esters in rats. J Pharm Sci 61:51-55.

Singh AR. Lawrence WH, Autian J. 1975. Maternal-fetal transfer  of
DEHP and 14C-diethyl phthalate in rats. J  Phann Sci 64:1347-1350.

SJoberg P. Bondesson U, Gray TJ, Ploen L.  1986a. Effects  of DEHP and
five of its metabolites on rat testis  in vivo  and in vitro. Arch Toxicol
58:72-77.

* SJoberg P, Lindquist NG,  Ploen L. 1986b. Age -dependent  response of the
rat testes to DEHP. Environ Health Perspect 65:237-242.

SJoberg POJ, Bondesson UG,  Sedin EG, Gustaffson JP. 1985a. Exposure of
newborn infants  to plasticizers: Plasma  levels of DEHP and MEHP during
exchange transfusion. Transfusion 25:424-428.

SJoberg P, Bondesson U, Hammarlund M.  1985b.  Nonlinearities in the
pharmacokinetics of DEHP and metabolites  in male rats. Arch Toxicol
58:72-77.

SJoberg P, Bondesson U, KJeilen L, Lindquist NG, Montin G, Ploen L.
1985c. Kinetics  of DEHP  in Immature and mature  rats and effect on
testis. Acta Pharmacol Toxicol  56:30-37.

SJoberg POJ, Bondesson UG, Sedin EG, Gustaffson JP. 1985d. Disposition
of DEHP and MEHP in newborn  infants subjected  to exchange  transfusions.
Eur J Clin Invest  15:430-436.

SRI  (SRI International). 1987.  Directory of Chemical  Producers.  Menlo
Park, CA: SRI  International.

Staubll W, Hess R.  1975. Lipoprotein formation in  the liver  cell.
Ultrastructural and  functional aspects relevant to hypolipidemic action.
In: Kritchersky D, ed.  Handbook of  Experimental Pathology.  Vol.  41.
Hypolipidemic/Agents.  Berlin:  Springer-Verlag:  pp. 229-289.

Sternlieb  I.  1979.  Electron  microscopy of  mitochondria and peroxisomes
of human hepatocytes.  In:  Popper  H. Schaffner  F.  eds. Progress in  Liver
Diseases. Vol.  6.  New York:  Grune  and Gratcon.  pp. 81-104.

-------
                                                        References    LI.

 Svoboda  DJ, Azarnoff DL  1979. Tumors  in male rats  fed echyl
 chlorophenoxy-isobucyrace, a hypolipidemic drug  Cancer Res  39 34L9-
 3428.

 Teirlynck OA,  Belpaire F  1985. Disposicion of orally administered DEH?
 and MEHP in the rat. Arch Toxicol 57.226-230.

 Thiess AM, Fleig I. 1979. Chromosomen untersuchungen bei micarbeicern
 tnic exposition gegenueber Di-2-aethylhexylphthalat (OOP). Berichtigurg
 Zbl Arbeitsmed. Bd. 29, H. 4, April 1979, S. 120, pp. 351-355

 Thiess AM, Korte A, Fleig H.  1978a. Untersuchengen zur morbidicac bei
 Mitarbeitern rait exposition gegenueber Di-2-aethylhexylphchalac (OOP)
 Vortr Anl d Jahrestg d Deutschen Gesellschaft f Arb Med in Frankfurt: V
 25. - 27.5 78. pp. 137-151.

 Thiess AM, Frentzel-Beyme R,  Wieldand R. 1978b. Mortality study in
 workers  exposed to di-(2-ethylhexyl)phthalate (OOP) (German). In:
 Moglichkerten and Grenzen des Biological Monitoring.  Arbeitsmedizini.sche
 Probleme des Dienstleistungsqewerbes.  Arbeitsmedizinisches kolloquium
 [Possibilities and Limits of Biological Monitoring. Problems of
 Occupational Medicine in Small Industries. Colloquium in Occupational
 Medicine], Frankfurt/M.,  May 1978. Stuttgart: AW Centner, pp. 155-164
 (cited in IARC 1981).

 Thomas GH. 1973.  Quantitative determination and confirmation of idencirv
 of trace amounts of dialkyl  phthalates in environmental samples  Enviror
 Health Perspect 3:23-28.

 Thuren A. 1986. Determination of phthalates in aquatic environments
 Bull Environ Contain Toxicol  36:33-40.

 Tomaszewski KE, Agarwal DK,  Melnick RL. 1986. In vitro steady-state
 levels of hydrogen peroxide  after exposure of male F344 rats and female
 B6C3F1 mice to hepatic peroxisome proliferators. Carcinogenesis 7 187L-
 1876.  .

 Tomita I, Nakamura Y,  Aoki N, Inui N.  1982. Mutagenic/carcinogenic
 potential of DEHP and MEHP.  Environ Health Perspect 45:119-125.

Tomlca I, Nakamura Y,  Yagi Y. Tutikawa K. 1986. Fetotoxic effects of
MEHP in mice.  Environ Health Perspect 65:249-254.

 Trebbln WM. 1979.  Hemodialysis and pregnancy. J Am Med Assoc 241.1811-
 1812.

Turnbull D. Rodricks JV.  1985.  Assessment of possible carcinogenic risk
 to humans resulting from exposure to DEHP. J Am Coll Toxicol 4-111-146

USDHHS (U.S.  Department of Health and Human Services). 1985. Fourth
Annual Report on Carcinogens. Summary 1985. Public Health Service.
Washington, DC: U.S.  Department of Health and Human Services, pp. 83-85

-------
112   Section 10

Verschueren K. 1977. Handbook of Environmental  Data on Organic
Chemicals. New York. NY:  Van Nostrand Reinhold.

Verschueren K. 198.. Handbook of Environmental  Data on Organic
Chemicals. New York. NY:  Van Nostrand Reinhold.

Vogel EW  1985. The drosophila somatic recombination and mutation assay
(SRM) using the white-coral somatic eye color systems.  Prog Mutat Res
5:313-317.

* Von Halle ES. 1985. A tabular review of the published mutagenicity
literature for IPCS study compounds.  Prog Mutat Res 5:699-725.

Ualdock MJ. 1983. Determination of phthalate esters in samples from the
marine environment using GC-MS. Chem Ecol 1:261.

Walseth F. Toftgard R, Nilsen OG. 1982. Phthalate esters. I: Effects on
cytochrome P-450 mediated metabolism in rat liver and lung, serum
enzymatic activities, and serum protein levels. Arch Toxicol 50'1-10.

Ward JM, Diwan BA, Ohshimna M. Hu H.  Schuller HM, Rice JM. 1986. Tumor-
initiating and promoting activities of DEHP in vivo and in vitro.
Environ Health Perspect 65:279-291.

Ward JM, Rice, JM. Creasia D, Lynch P. Riggs C. 1983. Dissimilar
patterns of promotion by DEHP and phenobarbital of hepatocellular
neoplasia initiated by diethylnitrosamine in B6C3F1 mice. Carcinogenesis
4-1021-1030.

Warf Institute. 1976. Acute  inhalation LC50 Sample LL-1132. Unpublished
study. July 23.

Warren JR, Lalwani ND. Reddy JK. 1982. Phthalate esters as  peroxisome
proliferator  carcinogens. Environ Health Perspect 45:35-40.

Watts P.  1985. Di-2-ethylhexylphthalate metabolism  in man.  Food  Chem
Toxicol 23:1023.

White RD, Carter  DE,  Earnest 0,  Mueller J.  1980. Absorption and
metabolism of three  phthalate  diesters by the  rat  small  intestine   Food
Cosmet Toxicol 18:383-386.

Williams  DT.  1973.  Dibutyl-  and di-(2-ethylhexyl)phthalate in fish. J
Agric Food Chem  21:1128-1129.

Williams  DT.  Blanchfield  BJ.  1974. Retention,  excretion,  and  metabolism
of  DEHP administered orally  to the rat.  Bull Environ Contain Toxicol
11:371-387.

Williams  CM.  Tong C.  Brat SV.  1985.  Tests with the rat hepatocyte
primary culture/DNA-repair  test.  Prog Mutat Res  5.341-345.

Wolfe NL  et  al.  1980.  Chemosphere  9  393-402 (cited in HSDB 1987).

-------
                                                        References
* Wolkowskl-Tyl R. Jones-Price C, Marr MC. 1984. Teratologlc evaluation
of dlethylhexyl phthalate (CAS No. 117-81-7) In Fischer 344 rats
Report: ISS RTI-60, FDA/NCTR-84/135.  Order PB85-105658/GAR, p. 224

Wolkowski-Tyl R.  Jones-Price C.  Marr MC. Kirarael CA. 1984b. Teratologic
evaluation of diethylhexyl phthalate in CD-I mice. Final report.
National Center for Toxicological Research,  Jefferson, AR. PB85-105674

Wurgler FE. Graf U, Frei H.  1985. Somatic mutation and recombination
test in wings of Drosophila  melanogascer. Prog Mutat Res 5:325-340

-------
                                                                     LL5
                             11.   GLOSSARY

Acute Exposure--Exposure to a chemical for a duration of 14 days or
less, as specified in Che Toxicological Profiles.

Bioconcentration Factor (BCF)--The quotient of the concentration of a
chemical in aquatic organisms at  a specific time or during a discrete
time period of exposure divided by the concentration in the surrounding
water at the same time or during  the same time period.

Carcinogen--A chemical capable of inducing cancer.

Ceiling value (CL)--A concentration of a substance that should not be
exceeded, even instantaneously.

Chronic Exposure--Exposure to a chemical for 365 days or more, as
specified in the Toxicological Profiles.

Developmental Toxicity--The occurrence of adverse effects on the
developing organism that may result from exposure to a chemical prior to
conception (either parent), during prenatal development, or postnatally
to the time of sexual maturation. Adverse developmental effects may be
detected at any point in the life span of the organism.

Embryotoxicity and Fetotoxicity--Any toxic effect on the conceptus as a
result of prenatal exposure to a  chemical; the distinguishing feature
between the two terms is the stage of development during which the
insult occurred. The terms, as used here, include malformations and
variations, altered growth, and In utero death.

Frank Effect Laval (FED--That level of exposure which produces a
statistically or biologically significant increase in frequency or
severity of unmistakable adverse  effects, such as irreversible
functional impairment or mortality, in an exposed population when
compared with its appropriate control.

EPA Health Advisory—An estimate  of acceptable drinking water levels for
a chemical substance based on health effects information. A health
advisory is not a legally enforceable federal standard, but serves as
technical guidance to assist federal, state, and  local officials.

Immediately Dangerous to Life or  Health (IDLH)--The maximum
environmental concentration of a  contaminant from which one could escape
within 30 min without any escape-impairing symptoms or  irreversible
health effects.

-------
116   Section 21

Intermediate Exposure--Exposure  to  a  chemical  for  a  duration of 15-364
days,  as specified in the Toxicological  Profiles

ImmunoLoglc Toxlcity--The occurrence  of  adverse effects  on the  immune
system that may result  from exposure  to  environmental  agents such  as
chemicals

In vitro--Isolated from the living  organism and artificially maintained.
as in a test tube

In vivo--Occurring within the living  organism.

Key Study--An animal or human toxicological study  that best illustrates
the nature of the adverse effects produced and the doses associated with
those effects.

Lethal Concentration(LO) (LCLO)--The  lowest concentration of a chemical
in air which has been reported to have caused death in humans or
animals

Lethal Concentration(SO) (LCSO)--A calculated concentration of a
chemical in air to which exposure for a specific  length of  time is
expected to cause death  in 50% of a defined experimental animal
population.

Lethal Dose(LO) (LDLO)--The lowest dose of a chemical introduced by a
route other than  inhalation that is expected to have caused death  in
humans or animals

Lethal Dose(50) (LDSO)--The dose of a chemical which has been  calculated
to cause death  in 50% of a defined experimental animal population.

Lowest-Observed-Adverse-Effect Level  (LOAEL)--The lowest dose  of
chemical  in a study  or  group of  studies which produces statistically or
biologically  significant increases in frequency or  severity of adverse
effects between the  exposed population and  its appropriate  control.

Lowest-Observed-Effect  Level (LOEL)--The  lowest dose  of chemical  in a
study  or  group  of studies  which  produces  statistically or biologically
significant  increases  in frequency or severity of effects between the
exposed  population  and  its appropriate control.

Malformations--Permanent structural  changes  that  may  adversely affect
survival,  development,  or  function.

Minimal  Risk  Level — An  estimate  of daily  human exposure  to  a chemical
that  is  likely  to be without an  appreciable risk  of deleterious effects
(noncancerous)  over a  specified  duration  of exposure.

Mutagen--A substance that causes mutations.  A mutation is a change in
the  genetic  material in a body  cell.  Mutations can lead to birth
defects,  miscarriages,  or cancer.

-------
                                                          Glossary   117

Neurotoxicity--The occurrence of adverse effects on the nervous system
following exposure to a chemical.

No-Observed-Adverse-Effect Level (NOAEL)--That dose of chemical at which
there are no statistically or biologically significant increases in
frequency or severity of adverse effects seen between the exposed
population and its appropriate control  Effects may be produced at this
dose, but they are not considered to be adverse.

No-Observed-Effect Level (NOEL) --That dose of chemical at which there
are no statistically or biologically significant increases in frequency
or severity of effects seen between the exposed population and its
appropriate control.

Permissible Exposure Limit (PEL) --An allowable exposure level in
workplace air averaged over an 8-h shift.

q, --The upper-bound estimate of the low-dose slope of the dose- response
curve as determined by the multistage procedure. The q * can be used co
calculate an estimate of carcinogenic potency, the incremental excess-
cancer risk per unit of exposure (usually /*g/L for water, mg/kg/day for
food, and Mg/m^ for air).
Reference Dose (RfD)--An estimate (with uncertainty spanning perhaps an
order of magnitude) of the daily exposure of the human population to a
potential hazard that is likely to be without risk of deleterious
effects during a lifetime. The RfD is operationally derived from the
NOAEL (from animal and human studies) by a consistent application of
uncertainty factors that reflect various types of data used to estimate
RfDs and an additional modifying factor, which is based on a
professional judgment of the entire database on the chemical. The RfDs
are not applicable to nonthreshold effects such as cancer.

Reportable Quantity (RQ)--The quantity of a hazardous substance that is
considered reportable under CERCLA. Reportable quantities are: (1) 1 Ib
or greater or (2) for selected substances, an amount established by
regulation either under CERCLA or under Sect. 311 of the Clean Water
Ace. Quantities are measured over a 24 -h period.

Reproductive Toxic ity- -The occurrence of adverse effects on the
reproductive system that may result from exposure to a chemical. The
toxicity may be directed to the reproductive organs and/or the related
endocrine system. The manifestation of such toxicity may be noted as
alterations in sexual behavior, fertility, pregnancy outcomes, or
modifications in other functions that are dependent on the integrity of
this system.

Short-Term Exposure Limit (STEL)--The maximum concentration to which
workers can be exposed for up to 15 min continually. No more  than four
excursions are allowed per day, and there must be at least 60 min
between exposure periods. The daily TLV-TWA may not be exceeded.

-------
118   Section 11

Target Organ Toxlcity--This term covers a broad range of adverse effects
on target organs or physiological systems (e.g.,  renal,  cardiovascular)
extending from those arising'through a single limited exposure to those
assumed over a lifetime of exposure to a chemical.

Teratogen--A chemical that causes structural defects that affect che
development of an organism.

Threshold Limit Value (TLV)--A concentration of a substance to which
most workers can be exposed without adverse effect. The  TLV may be
expressed as a TWA, as a STEL, or as a CL.

Time-weighted Average (TWA)--An allowable exposure concentration
averaged over a normal 8-h workday or 40-h workweek.

Uncertainty Factor (UF)--A factor used in operationally  deriving the RfD
from experimental data. UFs are Intended to account for  (1) the
variation in sensitivity among the members of the human  population,
(2) the uncertainty in extrapolating animal data to the  case of humans,
(3) the uncertainty in extrapolating from data obtained in a study that
is of less than lifetime exposure, and (4) the.uncertainty in using
LOAEL data rather than NOAEL data. Usually each of these factors is set
equal to 10.

-------
                                                                      119
                         APPENDIX:  PEER REVIEW

     A peer review panel was assembled for DEHP. The  panel  consisted of
the following members: Dr. W. H. Lawrence, University of Tennessee.
Dr. G. Michalopoulos, Duke University; and Dr. G. M.  Pollack. University
of North Carolina. These experts collectively have knowledge of DEHP's
physical and chemical properties, toxicokinetics. key health end poincs.
mechanisms of action, human and animal exposure, and  quantification of
risk to humans. All reviewers were selected in conformity with the
conditions for peer review specified in the Superfund Amendments and
Reauthorization Ace of 1986, Section 110.

     A joint panel of scientists from ATSDR and EPA has reviewed the
peer reviewers' comments and determined which comments will be included
in the profile. A listing of the peer reviewers' comments not
incorporated in the profile, with a brief explanation of the rationale
for their exclusion,  exists as part of the administrative record for
this compound.  A lisC of databases reviewed and a list of unpublished
documents cited are also included in the administrative record.

     The citation of the peer review panel should not be understood co
imply their approval of the profile's final content.  The responsibility
for the content of this profile lies with the Agency  for Toxic
Substances and Disease Registry.
     *US COVaNMOtrnUNTlNCOmCI.! tl« -13 ••>•<•«/    UCIONNO4

-------