1,4-DICHLOROBENZENE
                                              Q
                                             -I
                                             I-
                                             <* Q.
Agency for Toxic Substances and Disease Registry
U.S. Public Health Service

-------
                                                       ATSDR/TP-88/14
           TOXICOLOGICAL PROFILE FOR
               1,4-DICHLOROBENZENE
            Date Published — January 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. 18S7-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 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  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.
                                                                     ILL

-------
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.
                                    f~-
                                    James 0.  Mason,  M.D.,  Dr.  P.H.
                                    Assistant Surgeon General
                                    Administrator, ATSDR
iv

-------
                                CONTENTS

FOREWORD 	  iii

LIST OF FIGURES 	   ix

LIST OF TABLES	   xi

 1.   PUBLIC HEALTH STATEMENT 	    1
     1.1  WHAT IS p-DICHLOROBENZENE? 	   1
     1.2  HOW MIGHT I BE EXPOSED TO p-DICHLOROBENZENE?  	   1
     1.3  HOW DOES p-DICHLOROBENZENE GET INTO MY BODY?  	   1
     1.4  HOW CAN p-DICHLOROBENZENE AFFECT MY HEALTH?  	   2
     1.5  IS THERE A MEDICAL TEST TO DETERMINE IF I  HAVE  BEEN
          EXPOSED TO p-DICHLOROBENZENE? 	   2
     1.6  WHAT LEVELS OF EXPOSURE HAVE RESULTED IN HARMFUL
          HEALTH EFFECTS? 	   2
     1.7  WHAT RECOMMENDATIONS HAS THE FEDERAL GOVERNMENT
          MADE TO PROTECT HUMAN HEALTH? 	   6

 2.   HEALTH EFFECTS SUMMARY 	   7
     2.1  INTRODUCTION 	   7
     2.2  LEVELS OF SIGNIFICANT EXPOSURE 	   8
          2.2.1  Key Studies 	   8
                 2.2.1.1  Lethality/decreased longevity  	   14
                 2.2.1.2  Systemic/target organ toxicity  	   14
                 2.2.1.3  Developmental toxicity 	   16
                 2.2.1.4  Reproductive toxicity 	   17
                 2.2.1.5  Genotoxicity 	   17
                 2.2.1.6  Carcinogenicity 	   18
          2.2.2  Biological Monitoring as a Measure  of
                 Exposure and Effects 	   20
          2.2.3  Environmental Levels as Indicators  of
                 Exposure and Effects 	   20
     2. 3  ADEQUACY OF DATABASE 	   20
          2.3.1  Introduction 	   20
          2.3.2  Health Effect End Points 	   21
                 2.3.2.1  Introduction and graphic summary 	   21
                 2.3.2.2  Descriptions of highlights of graphs  	   24
                 2.3.2.3  Summary of relevant ongoing research  ....   24
          2.3.3  Other Information Needed for Human
                 Health Assessment 	   25
                 2.3.3.1  Pharmacokinetics and mechanisms
                          of action 	   25
                 2.3.3.2  Monitoring of human biological  samples  ..   25
                 2.3.3.3  Environmental considerations  	   25

 3.   CHEMICAL AND PHYSICAL INFORMATION 	   27
     3.1  CHEMICAL IDENTITY 	   27
     3.2  CHEMICAL AND PHYSICAL PROPERTIES 	   27

-------
Contents

 4.  TOXICOLOGICAL DATA 	   31
     4.1   OVERVIEW 	[]['   31
     4 . 2   TOXICOKINETICS 	.' '_   3 L
           4.2.1   Overview 	   31
           4.2.2   Absorption 	    32
                  4.2.2.1  Inhalation  	   32
                  4.2.2.2  Oral  	   32
                  4.2.2.3  Dermal  	   32
           4.2.3   Distribution  	   32
                  4.2.3.1  Inhalation  	   32
                  4.2.3.2  Oral  	   33
           4.2.4   Metabolism	   34
                  4.2.4.1  Inhalation  	    34
                  4.2.4.2  Oral  	   34
                  4.2.4.3  Dermal  	   34
           4.2.5   Excretion 	   35
                  4.2.5.1  Inhalation  	   35
                  4.2.5.2  Oral  	   35
                  4.2.5.3  Dermal  	   35
           4.2.6   Discussion	            35
     4. 3   TOXICITY 	'.'.'.'.'.   36
           4.3.1   Lethality and Decreased Longevity 	   36
                  4.3.1.1  Overview  	   36
                  4.3.1.2  Inhalation  	                 36
                  4.3.1.3  Oral  	'   35
                  4.3.1.4  Dermal  	   38
                  4.3.1.5  Discussion  	   38
           4.3.2   Systemic/Target Organ Toxicity 	   38
                  4.3.2.1  Overview  	   33
                  4.3.2.2  Central nervous system effects  	   39
                  4.3.2.3  Liver effects 	   40
                  4.3.2.4  Renal effects 	   44
           4.3.3   Developmental Toxicity 	   46
                  4.3.3.1  Overview  	   46
                  4.3.3.2  Inhalation	     46
                  4.3.3.3  Oral  	   47
                  4.3.3.4  Dermal  	   47
                  4.3.3.5  Discussion  	   47
           4.3.4   Reproductive Toxicity 	   48
                  4.3.4.1  Overview  	   48
                  4.3.4.2  Inhalation  	   48
                  4.3.4.3  Oral  	\\\'m   43
                  4.3.4.4  Dermal  	   48
                  4.3.4.5  Discussion  	   48
           4.3.5   Genotoxicity 	   48
                  4.3.5.1  Overview  	   48
                  4.3.5.2  Microbial systems 	   49
                  4.3.5.3  Higher plants 	   49
                  4.3.5.4  Mammalian systems 	   49
                  4.3.5.5  Discussion  	   50
vi

-------
                                                               Contents

         4.3.6  Carcinogenicicy  	    50
                4.3.6.1  Overview  	   50
                4.3.6.2  Inhalation  	   50
                4.3.6.3  Oral 	   51
                4.3.6.4  Dermal  	   52
                4.3.6.5  Discussion  	   52
    4.4  INTERACTIONS WITH OTHER CHEMICALS 	   53

5.   MANUFACTURE, IMPORT, USE, AND DISPOSAL 	   55
    5.1  OVERVIEW 	   55
    5.2  PRODUCTION 	   55
    5.3  IMPORT 	    55
    5.4  USE 	    55
    5.5  DISPOSAL	   56

6.   ENVIRONMENTAL FATE 	   57
    6.1  OVERVIEW 	   57
    6.2  RELEASES TO THE ENVIRONMENT 	   57
         6.2.1  Anthropogenic Sources 	   57
         6.2.2  Natural Sources  	   58
    6.3  ENVIRONMENTAL FATE 	   58
         6.3.1  Atmospheric Fate Processes 	   58
         6.3.2  Surface Water/Groundwater Fate Processes 	   58
         6.3.3  Soil Fate Processes  	   59
         6.3.4  Biotic Fate Processes 	   59

7.   POTENTIAL FOR HUMAN EXPOSURE 	   61
    7.1  OVERVIEW 	   61
    7.2  LEVELS MONITORED OR ESTIMATED IN THE ENVIRONMENT 	   61
         7.2.1  Levels in Air 	   61
         7.2.2  Levels in Water  	   62
         7.2.3  Levels in Soil 	   64
         7.2.4  Levels in Food 	   64
         7.2.5  Resulting Exposure Levels 	   64
    7.3  OCCUPATIONAL EXPOSURES  	   65
    7.4  POPULATIONS AT HIGH RISK 	   67
         7.4.1  Above-Average Exposure 	   67
         7.4.2  Above-Average Sensitivity 	   67

8.   ANALYTICAL METHODS 	   69
    8.1  ENVIRONMENTAL MEDIA 	   69
         8.1.1  Air 	   69
         8.1.2  Water 	   71
         8.1.3  Soil 	   71
         8.1.4  Food 	   71
    8.2  BIOMEDICAL SAMPLES 	   71
         8.2.1  Fluids and Exudates  	   71
         8.2.2  Tissues 	   71

9.   REGULATORY AND ADVISORY STATUS 	   73
    9.1  INTERNATIONAL	   73
    9.2  NATIONAL	   73
         9.2.1  Regulations 	   73
         9.2.2  Advisory Guidance 	   76
         9.2.3  Data Analysis 	   77
                                                                    vli

-------
Concencs

     9.3  STATE 	    78
          9.3.1  Regulations 	   78
          9.3.2  Advisory Guidance 	   78

10.   REFERENCES 	   79

11.   GLOSSARY 	   91

APPENDIX:  PEER REVIEW 	   95
viii

-------
                            LIST OF FIGURES
1.1  Health effects from breathing p-DCB 	    3
1.2  Health effects from ingesting p-DCB 	    4
1.3  Health effects from skin contact with p-DCB 	    5
2.1  Effects of p-DCB--inhalation exposure 	    9
2.2  Effects of p-DCB--oral exposure 	   10
2.3  Effects of p-DCB--dermal exposure 	   11
2.4  Levels of significant exposure for inhalation of p-DCB  	   12
2.5  Levels of significant exposure for ingestion of p-DCB 	   13
2.6  Availability of information on health effects of p-DCB
     (human data) 	   22
2.7  Availability of information on health effects of p-DCB
     (animal data) 	   23
                                                                      ix

-------
                             LIST OF TABLES
3.1  Chemical identity of p-DCB 	   28
3.2  Physical and chemical properties of p-DCB 	    29
4.1  Reports of human exposure to p-DCB 	   37
7.1  Estimated human exposure to p-DCB by ingestion of
     drinking water 	   66
8.1  Analytical methods for p-DCB in environmental samples  	   70
8.2  Analytical methods for p-DCB in biological samples  	   72
9.1  Regulations and guidelines applicable to p-DCB 	   74

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

1.1  WHAT IS p-DICHLOROBENZENE?

     The compound 1,4-dichlorobenzene is most commonly referred to as
para-DCB or p-DCB, but there are approximately 20 additional terms used
for this chemical. Some of these names include Paramoth, para crystals,
and paracide, reflecting its widespread use as a moth killer. It is also
used to make deodorant blocks used in restrooms. At least 70 million
pounds of p-DCB are used each year in the manufacture of these two types
of products. It is also used in the manufacture of certain resins;
smaller amounts are used in the pharmaceutical industry and as a general
insecticide in farming.

     At room temperature, p-DCB is a colorless solid with a
characteristic penetrating odor. When exposed to air, it is slowly
transformed from its solid state into a vapor; the released vapor then
acts as a deodorizer and insect killer.

1.2  HOW NIGHT I BE EXPOSED TO p-DICHLOROBENZENE?

     p-Dichlorobenzene is released to the environment during both its
manufacture and use. Heavily populated and/or industrialized areas tend
to have the highest concentrations of p-DCB in air and in water,
including surface water, groundwater, and drinking water; whereas some
areas, especially rural areas, may have lower or undetectable levels.
Air is the major source of exposure to p-DCB.

     The persons most likely to be exposed to p-DCB are those who work
directly in its manufacture or processing, those who live in the
vicinity of any industrial area where it is produced, and some persons
who use it. People who live near chemical waste disposal sites may also
be subject to higher exposure due to local air or water contamination.
In addition, consumers are exposed to p-DCB through commonly used
household products, such as mothballs and deodorant blocks used in
public and household bathrooms. Thus, people can be exposed to p-DCB in
the air, via drinking water, or from handling products containing p-DCB.

1.3  HOW DOES p-DICHLOROBENZENE GET INTO MY BODY?

     The major way that p-DCB enters the body is through the lungs. For
example, this occurs when a person breathes p-DCB released from
industrial processes or from home use of p-DCB products.

     p-DCB can also get into our bodies when we drink water. p-DCB has
been found in drinking water in various locations throughout the United
States.

-------
 2   Section 1

      p-DCB may also enter our bodies  through  the  skin  if we  touch
 products that contain it,  such as  certain kinds of mothballs  and toilet
 deodorizer blocks.

      There is also  a possibility that p-DCB used  in the home  can be
 accidentally swallowed,  especially by young children. When p-DCB is used
 in mothballs or deodorant blocks,  these products  may be freely
 accessible in the closets or  bathrooms.

 1.4  HOW CAN p-DICHLOROBENZENE AFFECT MY HEALTH?

      There is no evidence that brief  low-level or moderate-level
 exposures to household products containing p-DCB  cause human health
 problems.  Higher p-DCB levels  in air, such as the levels that are
 sometimes associated with industrial  exposure, can cause headaches and
 dizziness.  Levels that would  result in death would be associated with an
 odor  so  intense  that it  would  be very unpleasant, if not intolerable,
 and would serve  as  a danger warning.  In industrial settings, workers
 exposed  to p-DCB at high levels are usually directed to wear
 respirators.

      In  laboratory  animals, breathing or ingestion of p-DCB can cause
 toxic effects  in the liver and kidney. Although there is no evidence
 that p-DCB can cause cancer in humans, laboratory animals treated with
 p-DCB in lifetime studies had  increased rates of cancer when compared
 with animals  not exposed to p-DCB.  Based on the results of these animal
 studies,  a potential exists that p-DCB may cause cancer in humans.  Based
 on  two animal  studies, there is some evidence that p-DCB exposure can
 result in birth  defects.

 1.5  IS  THERE A  MEDICAL  TEST TO DETERMINE IF I HAVE BEEN EXPOSED TO
     p-DICHLOROBENZENET

     A urine  test can determine if a person has been exposed to p-DCB
 and can  give a rough estimate  of the exposure level.  This test has  been
 used in  industrial  settings in surveys of occupational exposure. There
 is a specific compound (2,5-dichlorophenol)  that is produced in the body
 after exposure to p-DCB. Detection of this chemical in the urine will
 indicate  that  the person has been exposed to p-DCB within the previous
 day or two. The  amount of this  substance in urine is also a fairly  good
 indicator of the level of p-DCB in the air,  when air is the route of
 exposure. There  is  also  a blood test to measure levels of p-DCB in the
bloodstrean.

 1.6  WHAT LEVELS OF  EXPOSURE HAVE RESULTED IN HARMFUL HEALTH EFFECTS?

     The graphs on  the following pages (Figs.  1.1 and 1.2) show the
 relationship between exposure  to p-DCB and known health effects. In the
 first set of graphs,  labeled "Health effects from breathing p-DCB,*
exposure is measured in  parts of p-DCB per million parts of air (ppm).
 In the second set of graphs, the same relationship is represented for
the known "Health effects from  ingesting p-DCB." Exposures are measured
 in milligrams of p-DCB per kilogram of body weight per day (mg/kg/day).
Figure 1.3 shows known health effects in rats as a result of exposure to
p-DCB by skin contact. Exposure is  also measured in milligrams of p-DCB

-------
                                                     Public Health Statement
        SHORT-TERM EXPOSURE
    (LESS THAN OR EQUAL TO 14 DAYS)
 LONG-TERM EXPOSURE
(GREATER THAN 14 DAYS)
EFFECTS
IN
ANIMALS




CONG IN
AJR
(ppm)
1000



EFFECTS
IN
HUMANS
QUANTITATIVE
DATA WERE
NOT
AVAILABLE
EFFECTS CONC IN
IN AIR
ANIMALS (ppm)
1000



EFFECTS
IN
HUMANS
QUANTITATIVE
DATA WERE
NOT
AVAILABLE
BIRTH DEFECTS. — 800
REDUCED MATERNAL
WEIGHT GAIN
(RABBITS)

                600
                400
                                            LIVER AND
                                            KIDNEY EFFECTS-
                                            INRATS
                                            (1 5 YEARS)
               200
                150
                100
                50
         800
                                                              600
                                                               400
                                                              200
                                                              150
                                                              100
                                                               50
                     Fig. 1.1.  Health effects froa breatfaiag p-DCB.

-------
    Section
       SHORT-TERM EXPOSURE
   (LESS THAN OR EQUAL TO 14 DAYS)
 LONG-TERM EXPOSURE
(GREATER THAN 14 DAYS)
EFFECTS
IN DOSE
ANIMALS (mg/kg/day)
DEATH (RATS) 	 4000
DEATH (GUINEA 	
PIGS)
EFFECTS
IN
HUMANS
QUANTITATIVE
DATA WERE
NOT AVAILABLE
EFFECTS
IN DOSE
ANIMALS (mg/Vg/day)
4000
EFFECTS
IN
HUMANS
QUANTITATIVE
DATA WERE
NOT
AVAILABLE
               2000
                                                            2000
               1000
BIRTH DEFECTS—500
(RATS)           I
                                                            1000
         500
               300
               250
               200
               150
               100
               50
LIVER AND KIDNEY
EFFECTS (MICE
AND RATS)
         300
         250
                                                             200
                                                             150
         100
          50
                    Flf.1.2.  Hcaltfc rffcco tnm lagmiM p-DCB.

-------
                                                Public Health Scacemenc
        SHORT-TERM EXPOSURE
    (LESS THAN OR EQUAL TO 14 DAYS)
                                  LONG-TERM EXPOSURE
                                 (GREATER THAN 14 DAYS)
  EFFECTS
     IN
  ANIMALS

DEATH (RATS)
  DOSE
(mg/kg/day)
                6000
  EFFECTS
     IN
  HUMANS

QUANTITATIVE
DATA WERE
NOT AVAILABLE
  EFFECTS
    IN
  ANIMALS

QUANTITATIVE
DATA WERE
NOT AVAILABLE
  EFFECTS
    IN
  HUMANS

QUANTITATIVE
DATA WERE
NOT AVAILABLE
                Flf. 1J.  Heahfc effects froa akta cootect with p-DCB.

-------
 6    Section  1

 per  kilogram of body weight  per  day  (mg/kg/day). In all graphs, effects
 in animals are shown on the  left side, effects  in humans on  the right.
      These figures are  based only on studies in laboratory animals,
 usually mice and  rats.  Although  there have been several reports of
 harmful effects in humans  exposed to p-DCB, the levels and duration of
 exposure  are unknown.

      Decreased body weight gain  has  been observed in pregnant animals
 breathing p-DCB at similar levels (800 ppm) for a few days.  Slight
 abnormalities have also been observed in the circulatory systems of
 their offspring.

      The  results  of tests  in which rats and mice were administered p-DCB
 orally are shown  in Fig. 1.2. When small amounts (20 mg/kg) were
 administered, even for  a few days, subtle effects on the liver were
 seen. At  much higher levels  (over 1600 mg/kg),  a single dose resulted in
 the  death of test animals. With  long-term exposure, administering p-DCB
 orally at levels  of 150 mgAg/day led to damage to the liver and kidneys
 and,  at 300  mg/kg/day,  resulted  in cancer in these organs.

      Although there is  no  certainty  that the same health effects would
 occur if  humans were exposed to  p-DCB under the same conditions as the
 animals in these  studies,  scientists use the results in animals as a
 general guide to predict the potential toxicity of p-DCB in humans.
 Based on  the results of the  cancer studies in rats and mice, the excess
 risk of cancer in humans resulting from exposure to p-DCB has been
 calculated.  The number  of  excess cases of cancer in populations of
 10,000. 100,000,   1,000,000 or 10,000,000 individuals exposed to
 1 microgram  of p-DCB per liter (/*g/L) of drinking water for their entire
 lifetime  is  estimated to be  0.0063,   0.063, 0.63 and 6.3,  respectively.
 It should be noted that these risk values are upper-limit estimates.
Actual risk  levels are  unlikely  to be higher and may be lower.  It is
 also  important to note  that  in the case of p-DCB,  there is additional
 reason to view these values  as probably being overestimates of the
possible  cancer risk to  humans,  since the results that were reported for
 these test animals cannot be  directly related to human tissue or to the
usual route  and mode of human exposure.

 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 p-DCB. First,  the Environmental Protection Agency (EPA) has
 included p-DCB on the list of Hazardous Waste Constituents and subjected
 the  chemical to hazardous waste  regulations. Second, the EPA has issued
 test  rules requiring environmental and health effects testing of p-DCB.
Third, the EPA has promulgated a maximum contaminant level of
 75 micrograms per liter  of water for p-DCB in drinking water. Fourth,
all pesticides are registered with EPA,  and their manufacturers must
submit certain kinds of  information  to EPA in order that they be allowed
to be used as pesticides. Finally, the Occupational Safety and Health
Administration (OSHA) has a  maximum  permissible exposure limit to p-DCB
 in workplace air of 75 parts  per million.

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

 2.1  INTRODUCTION

      This  section summarizes and graphs  data on the  health  effects
 concerning exposure  to  p-DCB.  The purpose of this section is  to  present
 levels  of  significant exposure for p-DCB 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 p-DCB and (2)  a
 summarized depiction of significant exposure levels  associated with
 various adverse health  effects.  This  section also includes  information
 on  the  levels of p-DCB  that have been monitored in human fluids and
 tissues and information about  levels  of p-DCB  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 p-DCB 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 p-DCB.

-------
8   Section 2

2.2  LEVELS OF SIGNIFICANT 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, Ingestlon, and dermal--and then by toxicological end points
that are categorized Into six general areas--lethality, systemic/target
organ toxicity, developmental toxlcity, reproductive toxicity, genetic
toxicity, and carclnogenlcity. 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.
     Adjustment reflecting the uncertainty of extrapolating animal data
to man,  Intraspecles 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  thac  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  Kay Studies
      Figures 2.1 through 2.3  are graphic  presentations of the adverse
 health effects that have been identified in studies using inhalation and
 ingestion data, respectively.  None of the available human studies were
 considered acceptable.  There are various  limitations and deficiencies in
 these studies. For example,  neither the duration nor the intensity of
 exposure is clearly known or stated in any of these reports.  In
 addition,  it cannot be stated with certainty  that each of these persons
 was exposed to only p-DCB.  Therefore, only animal studies are designated
 as key studies for p-DCB. Studies using dermal exposure were not located
 in the available literature.
      Figures 2.4 and 2.5 present data for the various toxicity end
 points  for the inhalation and oral routes of exposure, respectively. The
 details of these studies are presented in the  following subsections. In
 addition to data on the NOAELs and LOAELs available from the various

-------
                                                         tfealch Effects  Summary   9
ANIMALS
1000
 100
  10
 OlL.
       • RABBITS. DEVELOPMENTAL EFFECTS. 13 DAYS
      (• RATS. UVEH AND KIDNEY EFFECTS 78 WEEKS
      \ • RATS DEVELOPMENTAL EFFECTS. 10 DAYS
       O RABBITS. DEVELOPMENTAL EFFECTS. 13 DAYS
       O RATS. UVER AND KIDNEY EFFECTS. 76 WEEKS
     • LOAEL FOR ANIMALS
O NOAEL FOR ANIMALS
                               HUMANS
                                (PP«n)
                                                                 1000
                                 r  QUANTITATIVE
                                    DATA WERE
                                    NOT AVAILABLE
                                                                  100
                              01
APPROXIMATE
ODOR THRESHOLD
                  Fig. 2.1. Effects of p-DCB—inhaUtkw

-------
10    Section 2
      ANIMALS
     (m(j/kfl/day)

    10.000 r-
                                                 HUMANS
    V.OOO
     100 L.
           (100% LETHALITY)



   • GUINEA PIGS (100% LETHALITY)


   • RATS. LQso


   O GUINEA PIGS  (0% LETHALITY)



-  ORATS  (0% LETHALITY)
             • MICE. UVER EFFECTS. 13 WEEKS
             • MICE. UVER TUMORS. 2 YEARS6
             • RATS. DEVELOPMENTAL EFFECTS. 10 DAYS

             O MICE. UVER EFFECTS. 13 WEEKS
         /f • RATS. KIDNEY EFFECTS. 13 WEEKS
         _ J • RATS. KIDNEY TUMORS. 2 YEARS
           1 • MICE. UVER AND KIDNEY EFFECTS. 2 YEARS
           lO RATS. UVER EFFECTS. 2 YEARS
         	O RATS. DEVELOPMENTAL EFFECTS. 10 DAYS
           f • RATS. KIDNEY EFFECTS, 2 YEARS
           IO RATS. KIDNEY EFFECTS. 13 WEEKS
                                                                QUANTITATIVE DATA
                                                                WERE NOT AVAILABLE
             LOAEL   ONOAEL
                       Flg.L2. Effects of j-DCB—oral exposure.

-------
                                             tfealch  Effects  Summary    11
   ANIMALS
  (mg/kg/day)
10,000
 1.000
  100
    10
   0.1
  0.01 •-
          RATS, LD5Q
HUMANS
       QUANTITATIVE
       DATA WERE
       NOT AVAILABLE
                   Fig. 2J. Effects of p-DCB—dermal exposure.

-------
12   Section 2
                 ACUTE
               (S14 DAYS)

             DEVELOPMENTAL
        INTERMEDIATE
        (15-364 DAYS)
  (ppm)

 10.000 r
  1.000
   100
     10
    0.1
   0.01
   0.001  L-
     QUANTITATIVE DATA
     WERE NOT AVAILABLE
          r RAT
          h RABBIT
|  MINIMAL RISK FOR EFFECTS
i  OTHER THAN CANCER
                 CHRONIC
                (>365 DAYS)

                 SYSTEMIC
                                                                r (LIVER)
                    • LOAEL
                    O NOAEL
             Fig. 2.4. LerebofdptflcaM
for
                          of fDCB.

-------
                                                         Healch  Effects  Summary    13
                      ACUTE
                    (S14 DAYS)
                     INTERMEDIATE
                     (15364 DAYS)
 CHRONIC
(>365 DAYS)
                      DEVELOP-   TARGET    REPRO-    TARGET   TARGET
          LETHALITY3   MENTAL    ORGAN   DUCTION   ORGAN   ORGAN    CANCER6
(mg/kg/day)

  10.000 r
   1.000
     100
      10
     01
    001 -
   0001 -
  00001 -
 000001 -
0 000001 L-
          r  RAT
          m MOUSE
          g  GUINEA PIG
                      r (KIDNEY) f m (LIVER)
                               O        i
                                                                           • m
                                                     I
                                        •  m (LIVER.
                                 r (KIDNEY)    KIDNEY>
                                                                 V

                                                                  i
                                                    ESTIMATED
                                                    UPPER-BOUND
                                                    HUMAN
                                                    CANCER
                                                    RISK LEVELS6
                                                                       io-4i
                                                                       10-5-
                                                                       io~6-
                                                                       io-7j
 J  MINIMAL RISK FOR EFFECTS   "f  MINIMAL RISK LEVEL ESTIMATED
 I  OTHER THAN CANCER         i   FROM INTERMEDIATE EXPOSURE DATA
J,                           sir

 • LOAEL
 O NOAEL
     'Available Irtwlliy daa ar» not eontidarvd ad*qua» tor ealeUaong an MRL.

     'Combined adenomas plua cananomat.

     'Further anelyM of me reaulta of the NTP (1987) bwa*My nae nwed quetdona at » tfw ratewanoe
      lor humana of tfie ooaerxed tumota In raa and mice.
                 FIg.U. Lercb of
                           forhferttaofp-DCB.

-------
14   Section 2

studies, these figures present estimates of minimal risk levels for each
of the durations of exposure where data permit.

     The estimation of minimal risk levels is based on the use of
uncertainty factors with  the results of acceptable studies. When a NOAEL
has been determined from  an adequate animal study of appropriate
duration, that NOAEL  is divided by an uncertainty factor of 100. For
example, if a NOAEL of 450 ppm has been determined for reproductive
effects in an adequate study using mice, a level of 4.5 ppm would be
considered to present minimal risk for this effect to humans.  If a LOAEL
has been used because a NOAEL was not available, an uncertainty factor
of 1000 is used. These uncertainty factors are intended to account for
interspecies variation and intrahuman sensitivity.
2.2.1.1  Lethality/decreased longevity
     No animal or human studies provided adequate data to evaluate
lethality or decreased longevity as a toxicity end point for p-DCB
exposure via inhalation.  Deaths reported in animal studies (and in human
case studies) with exposure to p-DCB via inhalation have been attributed
to massive renal or hepatic damage.
     One study on the lethality of p-DCB administered by gavage to rats
(groups of 5 to 15, mixed sex) and guinea pigs (groups of 5,  mixed sex)
was reported by Hollingsworth et al.  (1956).  In rats,  a single dose of
1000 mg/kg resulted in no lethality,  whereas  4000 mg/kg resulted in 100%
lethality.  In guinea pigs, 1600 mg/kg resulted in no lethality, whereas
2800 mg/kg resulted in 100% lethality.  These  data are presented in Fig.
2.5.  Gaines and Linder (1986) reported that oral LJ>50 values for p-DCB
in adult rats were 3863 and 3790 mg/kg for males and females,
respectively.  In the same study,  these authors reported that the dermal
LD50 for p-DCB in rats was greater than 6000  mg/kg in both sexes.

2.2.1.2  Systemic/target  organ toxicity

     The organ systems designated as primary  toxicity end points for
p-DCB are the central nervous system (CNS), the liver, and the kidneys.

     Central nervous system effects.   CNS toxicity has been reported in
a few case studies of humans exposed to p-DCB at unspecified levels via
inhalation in the home and in the workplace.  Effects have included
headache, dizziness, nausea, clumsiness, and slurred speech.  However,
because of various limitations and deficiencies associated with these
studies, they can be viewed only as suggestive evidence that p-DCB
inhalation by humans can  result in CNS effects. Detailed descriptions of
these studies are presented in Sect.  4.3. There is no acceptable
evidence of CNS effects in animals exposed to p-DCB by inhalation.
     CNS effects have not been adequately confirmed in association with
ingestion of p-DCB by humans, and no data are available in test animals.
Also, dermal studies are  not available in any species.

     Hepatotozlcity.  Hepatic effects including degeneration,  necrosis,
porphyria,  and/or changes in enzyme levels have been reported in animal
studies by both oral and  inhalational exposure during short-term (as
brief as 3 days) or long-term (2 years) exposures. Evidence of liver
damage in humans exposed  by inhalation include jaundice, cirrhosis, and

-------
                                             Healch Effects Summary   15

acute yellow atrophy of Che liver. Dose-response data, however,  are not
available for humans. Hepatic effects have not been reported in humans
ingesting p-DCB, and dermal studies are not available for either humans
or animals.
     Hepatotoxicity. inhalation.  Hepatic effects in humans exposed to
p-DCB via inhalation for durations of a few months or more have been
reported in several case studies. However, there are no dose-response
data associated with these reports. A single study in animals provides
quantitative data for hepatic effects associated with long-term
inhalation exposure to p-DCB. Riley et al. (1980) reported a NOAEL of
75 ppm for increased liver weight and slight increases in urinary
coproporphyrin in rats exposed to p-DCB for 76 weeks. The LOAEL was
500 ppm for these effects. This study serves as the basis for the
minimum risk level shown in Fig. 2.4.
     Hepatotoxicity, oral.  No acceptable data are available on the
hepatic effects of short-term, intermediate, or chronic oral exposure of
humans to p-DCB. However, hepatic effects have been observed following
oral exposures of animals to p-DCB.
     Available data on intermediate exposure of test animals to p-DCB
via ingestion indicate that various adverse hepatic effects are observed
in 13-week mouse studies at doses of 600 mg/kg/day and higher. A NOAEL
of 338 mg/kg/day was identified.
     In a 2-year NTP (1987) bioassay of p-DCB, male F344/N rats were
dosed by gavage at ISO or 300 mg/kg/day and females at 300 and 600
mg/kg/day. Hepatic effects were not observed in rats. In the same study,
male and female B6C3F1 mice were dosed by gavage with p-DCB at 300 or
600 mg/kg/day. p-DCB increased the incidences of nonneoplastic liver
lesions in male and female mice, including alteration in cell size
(cytomegaly and karyomegaly), hepatocellular degeneration, and
individual cell necrosis. Therefore, the LOAEL for hepatic effects in
mice has been identified as 300 mg/kg/day.
     Hepatotoxicity, dermal.  There are no available data on hepatic
effects resulting from short-term, intermediate, or long-term dermal
exposure to p-DCB by humans or animals.
     Renal effects, inhalation.  No acceptable data are available on
renal effects following inhalation exposure of any duration by humans to
p-DCB. In a long-term animal study by Riley et al. (1980), increased
renal weights were reported  in rats exposed to p-DCB via inhalation for
76 weeks. The NOAEL for these effects was 75 ppm, and the LOAEL was
500 ppm.
     Renal effects, oral.  There are no available data to suggest that
renal toxicity is associated with short-term oral exposure to p-DCB by
humans or test animals. Likewise, there are no quantitative data
available on the renal effects of intermediate or chronic oral exposure
of humans to p-DCB. However, adverse renal effects have been reported in
several subchronic studies in animals exposed to p-DCB by gavage.
Hollingsworth et al. (1956)  reported a NOAEL of 18.8 mg/kg/day and a
LOAEL of 188 mg/kg/day for increased renal weight and cloudiness and
swelling of the renal tubular epithelium  in female rats administered
p-DCB for 6 months.

-------
16   Section 2

     In two NTP (1987) 13-week pilot studies, F344/N rats and B6C3F1
mice were dosed with p-DCB by gavage. In the first study, rats were
dosed with 300 to 1500 mg/kg/day. Because histologic changes were
observed in the kidneys of male rats at all doses, a second 13-week
study was performed at doses of 38, 75, 150, or 600 mg/kg/day.
     Renal tubular cell degeneration was observed in male rats receiving
300 mg/kg/day or more in the first study, but only slight changes were
seen at 300 mg/kg/day in the second study.  The kidney weight to brain
weight ratio was increased in male rats receiving doses of 600 mg/kg/day
or more. The blood urea nitrogen level was increased slightly in male
rats dosed with 900 mg/kg/day or more. Based on the combined results of
both studies, a LOAEL of 300 mg/kg/day and a NOAEL of 150 mg/kg/day were
identified in rats. This study serves as the basis for the minimum risk
level shown in Fig. 2.5.
     Charbonneau et al. (1987a,b) and Bomhard and Luckhaus (undated)
reported that p-DCB administered by gavage to Fischer-344 rats under a
wide variety of dosage regimens resulted in an increase in renal hyaline
droplet formation in males but not in females. The Charbonneau et al.
(1987a) study showed that, after administration of a single 500-mg/kg
dose of l^C-p-DCB, the radioactivity in the kidney cytosol was found to
be reversibly associated with the protein a-2ft-globulin.

     Renal effects have also been observed in one study of chronic oral
exposure to p-DCB. In the NTP (1987) bioassay, described above, male
rats exposed to p-DCB at 150 mg/kg/day and 300 mg/kg/day for 2 years
exhibited nephropathy, epithelial hyperplasia of the renal pelvis,
mineralization of the collecting tubules in the renal medulla, and focal
hyperplasia of the renal tubular epithelium. There were increased
incidences of nephropathy in female rats dosed with p-DCB at 300
mg/kg/day and 600 mg/kg/day, as compared with vehicle controls.
Therefore, the LOAEL for renal effects in rats was 150 mg/kg/day.
Because this was the lowest level tested in this phase of the study, no
estimate of the NOAEL in rats is available. p-DCB at both 300 mg/kg/day
and 600 mg/kg/day in this study also increased the incidences of
nephropathy in male mice and renal tubular degeneration in female mice.
Therefore, 300 mg/kg/day was the LOAEL for renal effects in mice.
     Renal effects, dermal.  There are no available data appropriate for
determining a significant level of human exposure for renal effects
resulting from short-term, intermediate, or long-term dermal exposure to
p-DCB by humans or animals.

2.2.1.3  Developmental toxicity

     There are limited data suggesting that p-DCB is a developmental
toxicant at levels of exposure that result in maternal toxicity.
     Inhalation.  Exposures of pregnant rabbits to p-DCB at 800 ppm
resulted in slight maternal toxicity and developmental effects (an
increased incidence of retroesophageal right subclavian artery) . Hodge
et al. (1977, as summarized in Loeser and Litchfield 1984) reported that
500 ppm (the highest dose tested) was the NOAEL for maternal toxicity
and fetotoxic and teratogenic effects in rats exposed to p-DCB on days 6
to 15 of gestation. Hayes et al. (1985) reported a NOAEL of 300 ppm for

-------
                                              Health Effects Summary   L7

 maternal  and developmental toxicity In rabbits.  There was  a significant
 increase  (P  < 0.05)  in the incidence of retroesophageal right  subclavian
 artery at 800 ppm.  The authors did not consider  this increase  in
 incidence to be  a teratogenic effect.  However,  this effect was reported
 as  an observed structural anomaly in the development of these  fetuses,
 therefore, 800 ppm  is  the LOAEL for this study which serves as the  basis
 for the minimum  risk level shown in Fig. 2.4.

      Oral.   In a gavage study using rats,  Giavini  et al.  (1986)  reported
 that p-OCB at levels of 500 mg/kg/day and above  (administered  on days
 6 through 15 of  gestation) resulted in retarded  maternal weight  gain and
 the presence of  an extra rib in the fetuses.  The latter effect was  not
 considered by the authors to be a teratogenic effect;  however, other
 teratologists consider any structural  anomaly to be a significant event
 in  the  development of  the organism.  Thus,  based  on a conservative
 approach, the NOAEL  was 250 mg/kg/day.  This study  serves as  the  basis
 for the minimum  risk level shown in Fig.  2.5.

      Dermal.   There  are no available dermal studies on the  potential
 developmental  effects  of p-DCB exposure in humans  or test  animals.

 2.2.1.4   Reproductive  toxicity

      There are no adequate data in  experimental  animals or humans to
 assess  the potential reproductive toxicity of p-DCB.

      In a short-term inhalation study by Anderson  and Hodge  (1976,  as
 reported  in  Loeser and Litchfield 1983), no reduction in reproductive
 performance  (as measured by the percentage of males successfully
 impregnating  females)  was  observed  when male mice  were  exposed to p-DCB
 at  levels up  to 450 ppm for 5  days  and  then mated  with virgin  females.
 In  order  to assess the  potential effects of a compound  on  reproductive
 performance,  however,  studies  of this kind are usually carried out
 during the entire period of spermatogenesis (approximately 9 weeks).
 Therefore, the conditions  of this study are not  considered adequate to
 test  this effect.

     Neither oral nor  dermal  studies have been conducted to assess  this
 effect in humans or in test animals.

 2.2.1.5  Genotoxlcity

     Based on the results  of genotoxic  studies in  bacteria and mammalian
 cells, p-DCB is generally  viewed as  nonmutagenic (NTP 1987). Genotoxic
 effects have been observed in  several studies of higher plants exposed
 to p-DCB.   Investigations  using microbial, animal,  and human systems and
 cells have all been negative with two exceptions.  One  is a single study
 in  fungus (Prasad and  Framer  1968,  Prasad  1970). The other is  an
unpublished study indicating  that DNA replication  is  increased in the
 liver tissue  of male and female mice and in the  kidney tissue  of male
 rats administered a single  oral dose of p-DCB (Steinmetz and Spanggord
 1987a,b).

-------
18   Section 2

2.2.1.6  Carcinogeniclty

     Inhalation.  There are no available data on the potential
carcinogenicity of p-DCB  in humans exposed via inhalation for any
duration of exposure. Neither are data available on short-term or
intermediate exposures of test animals.

     No evidence of carcinogenicity was observed by Riley et al. (1980,
as reported in Loeser and Litchfield 1983) in male or female SPF Alderly
Park Wistar-derived rats. SPF Alderly Park Swiss mice were also used in
this study, but, because of a high incidence of respiratory infections,
the results are not considered valid. Rats were exposed to p-DCB by
inhalation at 0, 75, or 500 ppm for 5 h/day, 5 days/week for 76 weeks.
As discussed in Sect. 4.2, Hawkins et al. (1980), using radiolabeled
p-DCB, showed that tissue levels of radioactivity are similar when
repeated oral doses of 250 mg/kg/day or repeated inhalation doses of
1000 ppm for 3 h/day are administered. However, the conditions of the
Riley et al. (1980) study may have resulted in considerably lower tissue
levels of p-DCB than those occurring in animals in the NTP (1987) gavage
study (dosed at 150 mg/kg/day and 300 mg/kg/day for male rats and 300
and 600 mg/kg/day for female rats and mice of both sexes).  Another
consideration is that the maximum tolerated dose (MTD) was not achieved
in the inhalation study. Based on the dosage levels, as well as the
less-than-lifetime duration of dosing, the results of the Riley et al.
(1980) study cannot be viewed as providing definitive evidence of the
noncarcinogenieity of p-DCB via the inhalation route.

     Oral.  There are no available data on the potential carcinogenicity
of p-DCB in humans exposed by ingestion for any duration of exposure. In
animals, only data on chronic exposure are available.

     p-DCB was carcinogenic in both rats and mice exposed to p-DCB for
2 years in the NTP (1987) carcinogenesis bioassay.  p-DCB was
administered by gavage to F344/N rats and B6C3F1 mice in groups of 50
animals per sex per dosage group. Hale rats received doses of 150
mg/kg/day and 300 mg/kg/day; female rats and mice of both sexes received
doses of 300 mg/kg/day and 600 mg/kg/day.

     In this study, p-DCB produced a dose-related increase in the
incidence of tubular cell adenocarcinomas of the kidney in male rats
[1/50 (2%); 3/50 (6%); 7/50 (14%)]. These malignant tumors are uncommon
in male F344/N rats; they have been diagnosed in only 4/1098 (0.4%) of
the corn oil gavage controls in previous NTP studies. In addition, one
tubular cell adenoma was observed in a high-dose male rat.  There were no
tubular cell tumors in dosed or vehicle control female rats. There was a
marginal increase in the incidence of mononuclear cell leukemia in dosed
male rats compared with that in either vehicle controls or historical
controls. Based on this rat study, p-DCB was found to be carcinogenic in
male rats because of the finding of renal tumors.

     In this study, p-DCB also increased the incidences of
hepatocellular carcinomas in high-dose male mice and of hepatocellular
adenomas in both high- and low-dose male and in high-dose female mice.
The increase in adenomas plus carcinomas was statistically significant
at the high dose but not at the low dose. Female control mice in this
bioassay had a substantially higher Incidence of liver tumors than did

-------
                                              Healch Effects Summary   19

 historical  controls.  Hepatoblastomas (a form of hepatocellular
 carcinoma)  were  observed in four high-dose male mice with other
 hepatocellular carcinomas but not in vehicle controls.  This rare
 malignant tumor  had not previously occurred in 1091 male  vehicle  control
 mice  in NTP studies.  An increase in thyroid gland follicular cell
 hyperplasia was  observed in dosed male  mice,  and there  was a marginal
 positive trend in  the incidence  of follicular cell adenomas of the
 thyroid gland in female mice.  Pheochromocytomas (tumors of chromaffin
 tissue  of the adrenal medulla or sympathetic preganglia)  (benign  and
 malignant,  combined)  of the adrenal gland occurred with a positive  trend
 in  dosed male mice,  and the incidence in the high-dose  group was
 significantly greater than in vehicle controls.  The incidences of
 adrenal gland medullary hyperplasia and focal hyperplasia of the  adrenal
 gland capsule were  also elevated in dosed male mice.

     Further analysis of the  results of the  NTP (1987)  bioassay has
 raised  certain questions as to the relevance  of the observed renal
 tumors  in male rats  and hepatic  tumors  in mice to  the potential
 carcinogenicity  of p-DCB in humans.  The observation that  kidney tumors
 are induced in male but not female rats in response to  exposure to
 certain chemicals has been the subject  of current  research.
 Toxicologists at the  Chemical  Industry  Institute of Toxicology (CUT)
 have hypothesized that the male  rat kidney is  susceptible  to  the
 induction of certain  tumors because it  contains  the protein cr-2j»-
 globulin, which has not been  found at significant  levels  in female rats
 or  in mice  or humans  (Charbonneau  et al.  1987a). They have  demonstrated
 that a-2/i-globulin in combination  with  certain components  of  petroleum
 enhances the formation of  hyaline  droplets in  the  proximal  convoluted
 tubules  of  male rats.  The  resulting cellular  damage  and cell
 proliferation are hypothesized to  result  in enhanced tumor  formation.
 This group  is currently conducting studies to  determine whether similar
 events  are  observed during the induction  of renal  tumors  in male  rats
 exposed  to p-DCB (personal  communication  from J. Swenberg,  CUT,  June 9.
 1987).  Based on these  considerations, it  is not clear how  to  interpret
 the renal tumors in evaluating the potential carcinogenicity  of p-DCB in
humans.  These tumors  have  not been used by EPA  to  estimate  the
carcinogenic risk for p-DCB.

     There has also been much  discussion  of the interpretation of the
 finding of hepatocellular  carcinomas  and  adenomas  in mice  in  the NTP
 (1987) study. There was  a higher than usual rate of  these  tumors  in
control female mice.  Because p-DCB has  not been demonstrated  to be
mutagenic in any of the  microbial  or  mammalian systems  tested, NTP
 (1987) has suggested  that  it may act  as a tumor promotor.  However, no
studies have been conducted to investigate this possibility.

     The EPA Office of  Drinking Water (EPA 1987a,  52FR25690 1987) has
placed p-DCB into Category  C, possible human carcinogen. This category
 is for substances with  limited evidence of oncogenic potential in animal
studies in  the absence  of human data.

     In an analysis of  the  NTP (1987) carcinogenicity data  (available in
1986 as the  galley draft),  Battelle and Crump  (1986) used  the liver
tumors in male mice and  the linearized multistage model to  calculate a

-------
20   Section 2

q * of 2.2 x 10"2 (mg/kg/day)"1. Based on this calculation,  oral doses
(ing/kg/day)'1 associated with upper-bound risks of 10"4,  10'5,  10'6.  and
ID"' would be 4.5 x 10'3, 4.5 x 10'4,  4.5 x 10"5,  and 4.5 x  10'6
mg/kg/day.
     Using the male rat kidney tumor data in the NTP (1987)  study with
p-DCB. Battelle and Crump (1986) report a q.* of 6 x 10'3 by the
linearized multistage model as well as by trie multistage-Weibull and
Crump's multistage models, taking time to death into account.  This q *
is lower than the q.* of 2.2 x 10"2 obtained with the male mouse liver
tumor data from the NTP (1987) study and is,  therefore,  considered the
less conservative estimate of risk.
     Dermal.  There are no available data on the potential
carcinogenicity of p-DCB via the dermal route.

2.2.2  Biological Monitoring as a Measure of Exposure and Effects
     As discussed in Sect. 4.1, Toxicokinetics, adipose  tissue,  kidneys,
and liver are the primary compartments into which p-DCB  is deposited
after exposure by animals. As discussed in Sect. 4.3, Toxicity,  the
major target organs of p-DCB appear to be the CNS, liver, and kidneys.
     p-DCB and/or its metabolites can be measured in various biological
tissues in order to confirm prior recent exposure. Examples  are:

  1.  measurement of p-DCB in blood;
  2.  measurement of p-DCB in adipose tissue; and
  3.  measurement of the p-DCB metabolite, 2,5-dichlorophenol,  and/or
      its conjugates in urine.
     There are currently no data available to assess a potential
correlation between the values obtained with these measurements and the
toxic effects observed in humans or test species.

2.2.3  Environmental Levels as Indicators of Exposure and Effects
     There are no available case studies or epidemiologic investigations
that suggest that levels of p-DCB found in environmental media are
associated with significant human exposure. The major route of exposure
for the general population is expected to be via inhalation, especially
in indoor air as a result of vapor released from commercial  products.
The available data suggest that the levels of p-DCB  in outside air are
relatively insignificant although the compound  is widespread. Levels  in
groundwater and surface water are also relatively low for the general
population. Therefore, exposure via drinking water does not pose a
significant health hazard for most individuals, but  could be significant
in localized contaminated areas.

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:

-------
                                             Healch Effaces Summary   21

    "(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 p-DCB. Such gaps are
identified for certain health effects'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 p-DCB 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 p-DCB will be developed by ATSDR,
NTP, and EPA in the future.

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. 2.6 and 2.7, 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 "probable human carcinogen" by both EPA
     and the International Agency for Research on Cancer (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.

-------
                                           HUMAN DATA
                    NJ

                    K>
 SUFFICIENT

INFORMATION*
                                                                                                                                    n
                                                                                                                                    rt
                                                                                                                                    No
                                                                                                              V     SOME

                                                                                                                 INFORMATION
                                                                                                           J
                                                                                                                      NO

                                                                                                                INFORMATION
                                                                                                  INHALATION
                                                                                             DERMAL
LETHALITY
               ACUTE     INTERMEDIATE    CHRONIC   DEVELOPMENTAL  REPRODUCTIVE  CAHCINOGENICIIY

                                              /    Toxicrrv       TOXICITY
                    SYSTEMIC TOXICITV
                        'Sufficient Information exists to meat at least one of the criteria for cancer or noncancer end points.
                               Fig. 2.6. Availability of iBformatioa oa health effects ot f-DCB (human data).

-------
                                         ANIMAL DATA
                                                                                                         V  SUFFICIENT
                                                                                                         'INFORMATION*
                                                                                                         V    SOME
                                                                                                           INFORMATION
                                                                                                       J
                                                                                                                NO
                                                                                                           INFORMATION
                                                                                                  ORAL
                                                                                             INHALATION
                                                                                         DERMAL
LETHALITY        ACUTE     INTERMEDIATE    CHRONIC   DEVELOPMENTAL  REPRODUCTIVE  CARCINOGENIC!!*
          Z -   - /    TOXICITY       TOXICITV
                    SYSTEMIC TOXICITV

                         "Sufficient Information exists to meet at least one of the criteria for cancer or noncancer end points.
8
»—
3-
n
in
0}
                            Fig. 2.7. Availability of information on health effects of p-DCB (animal data).

-------
24   Section 2

     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.

     For systemic/target organ effects, data adequacy have been plotted
for the system/target organ which is most sensitive to p-DCB.

2.3.2.2  Descriptions of highlights of graphs

     As shown in Fig. 2.6, human data for p-DCB are not considered
adequate in any of the categories presented. None of the available case
studies provided information on exposure levels associated with any of
the reported effects. In addition, exposure to p-DCB was not actually
verified in every report, nor was exposure to other possible causative
agents excluded. The case studies based on exposure in occupational
settings have provided some data for systemic toxicity resulting from
intermediate and short-term inhalation exposures to p-DCB.

     In the data based on animal studies, as shown in Fig. 2.7, only
lethality data are available via the dermal route. In the few acceptable
studies conducted via inhalation, the levels tested were considered to
be below the MTD and may not have been high enough to assess the effects
(chronic toxicity, carcinogenicity, and developmental toxicity) that
were being tested. No inhalation data were available to assess
lethality,  reproductive toxicity, or the systemic effects due to acute
or intermediate-duration exposures. In studies via the oral route, data
were considered adequate for systemic toxicity resulting from
intermediate and chronic exposures to p-DCB, with the liver found to be
the most sensitive target organ in most laboratory animals tested. The
carcinogenicity study is also considered to be adequate. Data on
lethality and developmental toxicity are not considered adequate. Oral
studies on developmental toxicity are considered adequate, but no oral
studies on reproductive toxicity have been located.

2.3.2.3  Summary of relevant ongoing research

     A search of the National Institutes of Health (NIH) Computer
Retrieval of Information on Scientific Projects (CRISP) database gave no
indication that there is current research being conducted relevant to
the toxicity of p-DCB. The CUT, however, has been investigating the
relevance of renal tumors found in rats exposed to p-DCB in the NIP
(1987) bioassay (personal communication, J. Swenberg, CUT,
June 9, 1987).  Recent information from the Chlorobenzene Producers
Association (CPA) submitted by Rautio (1988) indicates that current and
planned research activities for p-DCB include the following:

   • The p-DCB manufacturers are sponsoring a reproductive effects study
     in rats entitled "Two-generation reproductive sr dy of Inhaled
     para-dichlorobenzene in Sprague-Dawley CD rats."

   • CPA and several Japanese p-DCB manufacturers will soon initiate
     further pharmacokinetic research regarding p-DCB. The object of
     this research is to characterize the effects of route of
     administration and exposure level on the distribution, metabolism.
     and elimination of p-DCB in the rat and mouse.

-------
                                              Health Effects Summary   25

    •  In  response  to  a  Federal  Data  Call-In issued  pursuant to  the
      Federal  Insecticide,  Fungicide,  and Rodenticide Act, 7 U.S.C.
      Section  136  ec  seq.,  the  p-DCB manufacturers  have agreed  to sponsor
      the  following studies:  a  21-day  dermal  toxicity study (rat),
      chronic  toxicity  study  (nonrodent),  and a general metabolism study.

2.3.3  Other  Information Needed  for Human Health Assessment

2.3.3.1   Pharmacoki.netics  and  mechanisms  of  action

     The  mechanisms  of carcinogenicity for p-DCB are currently in
question. There is speculation by NTP (1987)  that  it acts as a tumor
promoter  rather than as an initiator  of  the  carcinogenic process.
Studies to examine tumor promotion  as an  end point would be useful in
assessing the potential cancer risks  from human exposure to p-DCB.

      It appears that the exposure levels  used in the Riley et al. (1980)
inhalation studies resulted  in much lower tissue levels of p-DCB than
were achieved in  the oral  studies by  NTP  (1987). The generally negative
or very mild physiologic effects observed during this and other
inhalation studies provide indirect support  for this observation.

     These findings  suggest  that more detailed pharmacokinetic data
would be  especially  useful in  assessing previous studies, and that any
further study via inhalation should probably  be conducted at much higher
exposure  levels, assuming  that they are physically possible to achieve.
Based on  available information, there are no  ongoing studies being
conducted to resolve these issues.

2.3.3.2   Monitoring  of human biological samples

     There are at least three  methods used to test for human exposure to
p-DCB. The method most commonly used  in industry is analysis of urine
for the metabolite 2,5-dichlorophenol. The other two methods involve the
analysis  of blood or adipose tissue for p-DCB itself. The utility of
these methods has not been clearly  delineated. Measurement of urinary
2,5-dichlorophenol correlates  approximately  (correlation coefficient not
calculated) with the levels  of exposure during certain p-DCB
manufacturing and packaging  activities.  There are no known ongoing
studies that would be useful in correlating  levels in biological samples
with exposure levels.

2.3.3.3  Environmental considerations

     Current methodologies to  assess  the  levels of p-DCB in the
environment are satisfactory.  The bioavailability of p-DCB present in
soil and water is not clearly  understood.  The consequences of its
adsorption to organic matter in these media may decrease its
bioavailability.

     There have been few studies done or  calculations made to predict
the degradation and  movement of p-DCB in  the environment over time. Mosc
predictions are based on available  information on other chlorinated
benzenes. There are  no known ongoing  studies  to address these
deficiencies.

-------
                                                                      27
                 3.  CHEMICAL AND PHYSICAL INFORMATION

3.1  CHEMICAL IDENTITY

     The chemical formula, structure, synonyms, and identification
numbers for p-DCB are listed in Table 3.1.

3.2  CHEMICAL AND PHYSICAL PROPERTIES

     Important physical and chemical properties of p-DCB are listed in
Table 3.2.

-------
28    Section  3
                                  Table 3.1. fVaical Mntfty
                                   Property
                     Chemical name

                     Synonyms
                     Trade names
                     Chemical formula

                     Wiswesser line Dotation

                     Chemical structure
          Value
1.4,-Dichlorobenzene

1.4-Dichloorbenzeen (Dutch)
1,4-Dichlor-B (German)
1.4-Diclorobenzene (Italian)
Benzene. 1.4-Dichloro
Benzene. /j-Dichloro
Dichlorobenzene, para, solid
p-Chlorophenyl chlonde
p-DCB
p-Dichloorbenzeen (Dutch)
p-Dichlorbenzol (German)
p- Dichlorobenzene
p-Dicblorobeazol
p-Diclorobenzene (Italian)
Parazene
PDB
Paradichlorobenzene
Paradichlorooenzol

Parodi
Pema-perazoi
Santochtor
Para crystal!
Paracide
Paradow
Paramoth
Paranuggeu
Evola

C.H.C1,

GRDG
                     Identification numbers
                       CAS Rejtitry No.
                       NIOSH RTECS No.
                       EPA Hazardous Waste No.
                       OHM-TADS No.
                       DOT/UN/NA/IMCO Shipping No.
                       STCCNa
                       Hizudoos Subsunccs Data Bank Na
                       National Cancer Institute Na
106-46-7
CZ 4550000
U072
8200116
UN 1592. IMCO 6.1
49411 28
523
C54955
                         Sourct: HSDB 1987.

-------
                            Chemical  and Physical Informacion
29
          T»M*3.0. Physical and chemical propertie* of r-DCB
Propeny
Molecular weight
Color
Physical state
Odor
Odor threshold, ppm
Melting point, "C
Boiling point, °C
Autoignition temperature, °F
Solubility
Water. 20 to 30° C. mg/L
Organic solvents
Density at 20° C, g/mL
Vapor density, air - 1
Partition coefTicients
Octanol/water (£„) log
Organic carbon (Kx) mL/g
Vapor pressure 20 to 30° C. mm Hg
Henry's law constant, atm-m'/mol
Refractive index, 60° C
Flash point (closed cap). °C
Flammable limits. ft
Value
14701
White or colorless
Crystalline solid
Aromatic, penetrating
018 to 30
S3 1
174 (sublimes at room
temperature)
775
Practically insoluble (79 mg/L)
Soluble in alcohol, acetone,
ether, chloroform, carbon
duulfide, and benzene
1 2475
5.08
360
1700
1 18
2.89 X 10~3
15283
65.6
6.2 to 16
References
Weast 1985
Verschueren 1977
Weast 1985
Clayton and
Clayton 1981
Amoore and
Hautala 1983,
HSDB 1987
Weast 1985
Weast 198S
NFPA 1978"
Verschueren 1977
Weasl 1985
Weast 1985
Sax 1979"
EPA 1986a
EPA 1986a
EPA I986a
EPA I986a
Weast 1985
Sax 1979"
NFPA 1978"
"Cited in HSDB 1987.

-------
                                                                      31
                         4.  TOXICOLOGICAL DATA
4.1  OVERVIEW
     p-DCB is highly lipid-soluble and is therefore assumed Co'be
absorbed by all exposure routes. Although quantitative absorption
studies are not available, p-DCB is assumed to be 100% absorbed when
administered orally, and about 30% absorbed via inhalation when exposure
persists from 1 to 3 h.
     The major route of elimination appears to be the urine.
2,5-Dichlorophenol has been demonstrated to be the major urinary
metabolite of p-DCB in both animals and humans.
     Laboratory studies on the toxicity of p-DCB have shown that the
major noncarcinogenic effects are CNS effects and hepatic and renal
toxicity. Developmental effects have also been demonstrated in both oral
and inhalation studies. There is evidence that p-DCB is carcinogenic in
rats and mice when administered by gavage, with liver and kidneys as the
target organs.

4.2  TOXICOKINETICS

4.2.1  Overview
     Quantitative absorption studies are not available for p-DCB. This
compound is assumed, however, to possess absorption characteristics
similar to those of benzene and the smaller chlorinated aliphatics and,
as such, would be 100% absorbed when administered orally, and about 30%
absorbed via inhalation when exposure persists from 1 to 3 h. The
potential for dermal absorption has not been addressed.
     Animal studies have demonstrated that p-DCB, once absorbed, is
highly concentrated in adipose tissue, with much lower levels in liver
and kidney. Detectable levels have also been reported in blood, lung,
heart, and brain. Levels in fat have been shown to persist at least five
days after exposure.
     2,5-Dichlorophenol has been demonstrated to be the major urinary
metabolite of p-DCB in both animals and humans. This metabolite is
eliminated principally as conjugates of glucuronic or sulfuric acids.
Some elimination in feces and expired air has been observed, and there
is also evidence of reabsorption in the enterohepatic circulation and
excretion in bile.

-------
32   Section 4

4.2.2  Absorption

     p-DCB is sparingly soluble in water but highly lipid soluble and
thus can be expected to cross most barrier membranes,  including those of
the placenta and brain (EPA 1987a).

4.2.2.1  Inhalation
     Human.  No data are available on the absorption of p-DCB by humans
exposed via inhalation.
     Animal.  No studies have been located which determine the
percentage of a dose of p-DCB absorbed following inhalation exposure by
animals. The EPA (1987a) estimated that 30% of an inhaled dose of p-DCB
is absorbed when exposure continues for longer than 1  to 3 h. Hawkins et
al. (1980) estimated that tissue levels of p-DCB are similar when female
rats either receive ten repeated oral doses of 250 mg/kg/day or inhale
1000 ppm for 3 h/day for 10 days.

4.2.2.2  Oral
     Human.  No studies have been located which determine the amount of
p-DCB absorbed by humans following oral exposure.
     Animal.  No studies have been located which determine the amount of
p-DCB absorbed by animals following oral exposure.

     Based on the absorption characteristics of benzene and the smaller
chlorinated aliphatics (Astrand 1975. Dallas et al.  1983). EPA (1987a)
has assumed that 100% of an oral dose of p-DCB is absorbed.

4.2.2.3  Dermal

     Human.  No studies have been located which determine the absorption
of p-DCB by humans following dermal exposure.
     Animal.  No studies have been located which determine the
absorption of p-DCB by animals following dermal exposure.

4.2.3  Distribution
     p-DCB is lipophilic and tends to be most highly concentrated in
tissues with high fat content. Available data indicate that adipose
tissue is the main depot for p-DCB in laboratory animals, with much
lower levels in liver and kidney.  p-DCB has also been found in human
breast milk and in human tissue high in fat content.

4.2.3.1  Inhalation
     Human.  There are no available studies which determine the tissue
distribution pattern of p-DCB in humans exposed via inhalation. The
compound has been found, however,  in human blood, fatty tissue, and
breast milk, presumably as a result of exposure via inhalation.

     p-DCB has been detected in human adipose tissue and blood.  In a
study of Tokyo residents, detectable levels of p-DCB were found in all
of 34 adipose tissue samples and all of 16 blood samples tested (Morita
et al. 1975, Morita and Ohi 1975)

-------
                                                  Toxicologies! Data   33

      EPA (1983b) presented the results of a database search for
 information on several chemicals identified in human milk. For p-DCB,
 samples were collected from 42 lactating women in five locations in the
 eastern United States. Measured values ranged from 0.04 to 68 /ig/mL with
 an average of 9.15
      EPA (1986d)  reported the results of a national survey of various
 volatile organic  compounds found in composites of human adipose tissue
 samples collected from persons living in the nine geographic areas  that
 compose the United States (within this survey).  The specimens
 (subcutaneous,  perirenal, or mesenteric tissue)  were collected from
 October 1981 through September 1982 (FY 1982)  and were  excised during
 surgery or as part of postmortem examinations.  For each geographic
 location,  three age groups were represented:  0 to 14 years,  15 to 44
 years ,  and 45 or  more years .

     Positive results were detected for p-DCB in these  composites in
 every category  of analysis,  with levels ranging  from 0.012 to 0.50  >ig/g
 wet  tissue.

     Animal.  Hawkins et  al.  (1980)  investigated the tissue  distribution
 of p-DCB in adult female  CFY rats following inhalation,  oral,  or
 subcutaneous  exposure.  The inhalation exposure was ten  consecutive  daily
 doses of 14C-p-DCB at 1000 ppm for 3  h/day.  Distribution patterns for
 all  routes  were similar to those observed by Kimura et  al.  (1979),  as
 described below,  with highest concentrations  measured in fat (up to 598
 Mg/g via inhalation)  and  next highest levels  in  kidneys  and  liver.
 Concentrations  in kidney  and liver were -8 and 5%,  respectively, of that
 found in adipose  tissue irrespective  of the  route of exposure.

 4.2.3.2  Oral

     Human.  There  are  no available data on  the  distribution of p-DCB in
 humans  following  oral exposure.

     Animal.  Kimura  et al.  (1979) determined  the distribution of p-DCB
 in the  tissues of fasted  rats following a single oral dose of 200 mg/kg-
 The  highest concentration of  p-DCB was  found  in  adipose  tissue at all
 sampling intervals  up to  120  h postexposure, and peak concentrations in
 fat  occurred at 12  h  postexposure  (>800 /*g/g  tissue). Kidney and liver
 showed  the next highest levels of  p-DCB,  with  peak concentrations of
 32 i*g/g  and 24 j*g/g occurring at 6 to 12 h. These concentrations were
 -4 and  3%, respectively,  of  the  concentrations found in  adipose tissue.
 Low  levels of p-DCB were  also found in  blood,  lung,  heart, and brain.
 Host of  the p-DCB in  all  tissues except for adipose  had  disappeared
 within 48 h after administration of the chemical.  Low levels of p-DCB
 were still detected in  the adipose tissue after  120  h.

     Hawkins et al. (1980)  investigated the tissue distribution of  p-DCB
 in adult female CFY rats  following oral administration of  50,  125,  250.
 375,  or  500 mg/kg/day by  gavage  for 10  days. Distribution  patterns  for
 all  routes of administration  used  (oral,  inhalation,  and subcutaneous)
were similar to those observed by  Kimura et al.  (1979),  as described
 above,  with the highest concentrations  measured  in fat and next highest
 (but much lower)  levels in kidney  and liver.

-------
 34   Section 4

 4.2.4  Metabolism

     Following oral administration to rabbits, the p-DCB is oxidized
 principally to the corresponding dichlorophenol, 2,5-dichlorophenol. A
 very high percentage of  this metabolite is eliminated as conjugates of
 glucuronic or sulfuric acids (Azouz et al. 1955, Williams 1959).
 Conjugated dichlorophenols also appear to be the principal metabolic
 products of the DCB isomers in humans (Hallowell 1959, Pagnatto and
 Walkley 1965).

     The mechanism of p-DCB oxidation to 2,5-dichlorophenol has not yet
 been thoroughly investigated. The metabolism of p-DCB could involve the
 formation of an arene oxide intermediate,  as has been proposed to occur
 in the oxidative metabolism of many halogenated aromatic hydrocarbons
 (Jerina and Daly 1974).  Preston et. al.  (1983) have  reported that the
 ring hydroxylation of 2,2,5,5-tetrachlorobiphenyl (a dimer of p-DCB)
 takes place without the  formation of an arene oxide  intermediate. p-DCB
 has not been shown to be mutagenic in microbial or mammalian systems, a
 result that may be viewed as further suggestive evidence that an arene
 oxide intermediate is not involved in its metabolism.

 4.2.4.1  Inhalation

     Human.  The dichlorophenols appear to be the principal metabolic
 product of the DCB isomers in humans.  In a survey of persons
 industrially exposed to p-DCB,  Pagnatto and Walkley  (1965) used the
 urinary metabolite, 2,5-dichlorophenol,  as a measure of exposure to
 p-DCB.

     Animal.  There are no available data on the metabolism of p-DCB in
 animals exposed via inhalation.

 4.2.4.2  Oral

     Human.  In a case of accidental ingestion of an unknown quantity of
p-DCB crystals by a 3-year-old boy, analysis of urine specimens yielded
 four unidentified phenols as well as 2,5-dichlorophenol. These were
 shown to be conjugated with glucuronic and sulfuric  acids (Hallowell
 1959).

     Animal.  Kimura et al. (1979) identified two sulfur-containing
metabolites of p-DCB in Che tissues,  blood, and urine of rats following
 a single 200-mg/kg oral dose or a series of 800-mg/kg/day doses over a
week. These metabolites were identified as 2,5-dichlorophenyl methyl
 sulfoxide and 2,5-dichlorophenyl methyl sulfone. Very small amounts of
 these two compounds were found (never more than 2 Mg/g tissue). Based on
 two peak blood levels of the latter compound,  the authors postulated
 that this substance might undergo enterohepatic reabsorption and
circulation.

4.2.4.3  Dermal

     Human.  There are no available data on the metabolism of p-DCB by
humans following dermal exposure.

     Animal.  There are no available data on the metabolism of p-DCB by
 animals following dermal exposure.

-------
                                                 Toxicological Data.   35

4.2.5  Excretion

     The limited data available indicate that urine is the major route
of excretion of p-DCB.

4.2.5.1  Inhalation

     Human.   No data are available on the excretion of p-DCB by humans
exposed via inhalation.

     Animal.  Hawkins et al. (1980) administered 14C-p-DCB to female CFY
rats for 10 days via inhalation at 1000 ppm for 3 h/day.  The amount of
l^C excreted in the expired air during 45 h after the tenth dose
represented a small proportion of the total ^C excreted, and, to the
surprise of the authors, was lower after inhalation (0.2%) than after
oral doses (1.0%) .
     In rats with cannulated bile ducts, no   C was detected in the
feces up to 24 h after a single dose, whereas, of the total ^C
recovered, 97.4% was eliminated in the urine at 5 days post treatment.

4.2.5.2  Oral
     Human.  No data are available on the excretion of p-DCB orally
administered to humans.
     Animal.  Hawkins et al. (1980) administered 14C -p-DCB to female CFY
rats for 10 days orally in rats with cannulated bile ducts at 250
mg/kg/day. Nine percent of the recovered ^C was excreted in the feces
during the 24 h following the last dose and presumably represents
unabsorbed material. In noncannulated rats, 97% of the recovered I4C was
eliminated in the urine within 5 days posttreatment. Approximately 1%
was recovered in expired air.

4.2.5.3  Dermal

     Human.  No data are available on the excretion of p-DCB by dermal ly
exposed humans.

     Animal.  No data are available on the excretion of p-DCB by
dermally exposed animals.

4.2.6  Discussion
     Experiments with laboratory animals show that p-DCB is absorbed via
oral or inhalation exposure and is distributed mainly to adipose tissue,
with some distribution to the liver and kidney and minor amounts found
in other organs. Absorbed p-DCB is metabolized mainly by oxidation to
the dichlorophenol , conjugated with glucuronic or sulfonic acid, and
rapidly eliminated, mainly in the urine. There are no data to indicate
that the route of exposure has any effect on the subsequent metabolism
and excretion of p-DCB.

     A review of the existing database suggests that additional
metabolic and pharmacokinetic studies are required  in order to actually
predict the rate of absorption of this compound when exposure occurs
orally, dermally, or via inhalation.  (Current estimates are based on
data from benzene and the smaller chlorinated aliphatics.) Because p-DCB

-------
36   Section 6

is commonly used as an ingredient in mothballs and toilet deodorant
blocks, it may be handled or inhaled by a significant portion of the
population on a regular basis and also has a high potential for
accidental ingestion. Thus, there is potential for human exposure by all
three routes.

4.3  TOXICITY

4.3.1  Lethality and Decreased Longevity

4.3.1.1  Overview
     Currently available data on the effects of p-DCB exposure by any
route on potential lethality or decreased longevity are extremely
limited. There are no epidemiological data for human exposure via
inhalation and no adequate laboratory data on animals via this route.
     Data on lethality via oral exposure are not extensive but do
suggest that this is an area of concern for this compound.

4.3.1.2  Inhalation
     Human.  There are no acceptable data available on the effects of
inhalation of p-DCB on lethality or decreased longevity in humans.
Available data on case reports from human studies are summarized in
Table 4.1. It is important to note that case studies, such as the ones
reported here, are to be interpreted with caution, since they reflect
incidents in which an individual has apparently been exposed to p-DCB,
and they assume that there has been no other exposure to a potentially
toxic agent(s). There is usually little or no verification of these
assumptions. These studies are not scientifically equivalent to
carefully designed epidemiologic studies or carefully controlled and
monitored laboratory experiments; therefore, they are to be considered
only as providing suggestive evidence that p-DCB may cause the reported
effects.
     Only one report of human death attributed to p-DCB exposure has
been located in the literature. A middle-aged couple died within months
of each other of acute yellow atrophy of the liver (confirmed at
autopsy) (Cotter 1953). Their home had been "saturated" with p-DCB
mothball vapor for a period of at least 3 to 4 months. Clinical symptoms
included severe headache, diarrhea, numbness, clumsiness, slurred
speech, weight loss (50 pounds in three months in the case of the
husband), and jaundice. The wife died within a year of the initial
exposure.
     Animal.  Acceptable lethality data are not available for animals
exposed to p-DCB by inhalation.

4.3.1.3  Oral
     Human.  There are no available data investigating the potential
lethality or decreased longevity associated with ingestion of p-DCB by
humans.

-------
                                                        Toxicological Data    37

Subject
62-year-old male




19-year-old female


60-year-old male



Wife of male
above



36-year-old female


34-year-old female


52-year-old mate


20-year-old mate
(+ 26 workmates)

52-year-old female



3-year^ld TIE Iff

19-year-old female




21-year-old
pregnant female


Exposure
p-DCB in bath-
room



Preparation of
p-DCB for 18
months
Heavy p-DCB
mothball vapor
10 home for
3 to 4 months
As above




p-DCB moth
killer ia house

Demonstnting
p-DCB-oontain-
ing products
p-DCB exposure
in fur storage
plant
p-DCB manufac-
ture, 1 to 7
months exposure
12- to 15-year
exposure to
p-DCB mothballs
ia house
Played with
p-DCB crystals
4-J pellets
of p-DCB
ingested daily
for 15 yean

Pica for p-DCB
toilet Mocka
first trimester
Tablt4.1. Resorts of sa
Symptoms
Asthenia, dizziness




Asthenia, dizziness.
weight lou

Headache, weight loss.
diarrhea, tarry
stools, numbness.
clumsiness
Headache, weight and
strength loss.
abdominal swelling.
jaundice

Periorbital swelling.
intense headaches.
profuse rhinitis
Tiredness, nausea.
headache, vomiting

Weakness, nausea.
blood, vomiting,
jaundice
Loss of weight.
ffuhmirtjoiii docrc&jc
of appetite
ff»tijhi piugusive
dyspnea, fatigue.
muooid sputnoi


black urine
Increased patchy
fijmmimhm Of Skin



None except those
commonly associated
with early pregnancy
SBU exposure to p-DCB
Clinical report
Light hyperchronuc
anemia, after 1
month, increase in
anemia and
bypogranulocytosis
Slight anemia.
reactional
hyperleukocytosis
Acute yellow
atrophy of the
liver (confirmed
by autopsy)
Acute yellow
atrophy of the
liver (confirmed
by autopsy),
splenomegaly
Exposure to p-DCB


Subacute yellow
atrophy and
cirrhosis of liver
Subacute yellow
atrophy of the
liver
MCtfiCOIOflODlACiDUl
and other blood
pathologies
Pulmonary
granalomatosis.
fjirml n • nsnn 11 • ftf
IOCW OtICPOIIi 
-------
38   Section 4

     Animal.  One study on Che lethality of p-DCB administered by gavage
to rats (groups of 5 to 15, mixed sex) and guinea pigs (groups of 5,
mixed sex) was reported by Hollingsworth et al.  (1956).  In rats, a
single dose of 1000 mg/kg resulted in no lethality,  whereas 4000 mg/kg
resulted in 100% lethality. In guinea pigs, 1600 mg/kg resulted in no
lethality, whereas 2800 mg/kg resulted in 100% lethality.  Gaines and
Linder (1986) reported that oral LD50 values for p-DCB in adult rats
were 3900 and 3800 mg/kg for males and females,  respectively.

4.3.1.4  Dermal

     Human.  There are no available data on the  potential  lethality or
decreased longevity associated with dermal exposure  of humans  to p-DCB.
     Animal.  Gaines and Linder (1986) reported  that the dermal LDSO for
p-DCB in rats was greater than 6000 mg/kg in both sexes.

4.3.1.5  Discussion
     The effects of p-DCB on longevity have not  been addressed in any of
the available studies. Although the available data suggest that exposure
to high doses of p-DCB via inhalation can result in lethality  in humans,
the data were not sufficient to establish this effect. However, in oral
studies in test animals, single doses of 2800 and 4000 mg/kg resulted in
100% lethality in guinea pigs and rats, respectively, whereas  1600 and
1000 mg/kg resulted in no lethality.  Oral LDso values of 3900  and 3800
mg/kg have also been reported for rats.

4.3.2  Systemic/Target Organ Tozicity

4.3.2.1  Overview

     The major target organs of p-DCB are the liver and kidneys and
probably the CNS. Adverse effects on these tissues are reported in most
of the available studies, in most species tested, and for  various
durations of exposure. Acceptable data from human studies  are  lacking;
available suggestive data from case reports are  summarized in  Table 4.1.

     CNS toxicity effects have been reported in  several case studies in
humans who were exposed to p-DCB for periods ranging from a few months
to several years, and in a short-term study in animals.  None of these
studies, however, provides quantitative data related to CNS effects.

     Hepatic effects have been demonstrated in several animal  studies
with exposures from 3 days to 2 years (lifetime  studies).  Observed
effects have ranged from porphyria and enzyme changes to liver
degeneration and necrosis. Effects in humans exposed for 3 months or
more included jaundice and yellow atrophy of the liver.

     Renal effects have been reported in animal  studies of 13  weeks or
longer and have ranged from increased kidney weights to epithelial
hyperplasia of the renal pelvis, focal hyperplasia of the renal tubular
epithelium, and mineralization of the collecting tubules in the renal
medulla. Renal effects were not reported in studies of shorter duration;
however, in most studies, renal effects were not being investigated.
(Levels of hepatic enzymes, for example, may have been the only study
parameter.)

-------
                                                 Toxicologies! Data   39

4.3.2.2  Central nervous system effects

     Overview.  The dichlorobenzenes in general are reported to produce
sedation and anesthesia in animals after acute oral or parenteral
administration. Relatively high doses are needed to produce these
effects. Acute poisoning is characterized by signs of disturbance of the
CNS including hyperexcitability, restlessness, and muscle spasms or
tremors. The most frequent cause of death is respiratory depression (EPA
1987a).

     Acceptable data to support observation of CNS effects are limited
in the case of p-DCB. Although CNS effects have been reported in the
case studies of a few persons exposed to p-DCB in their homes or
workplaces, there are no available estimates of the levels to which they
were exposed.  Also, as previously stated, there are various limitations
and deficiencies in these studies.

     Inhalation, human.  CNS effects have been reported in case studies
of humans exposed to p-DCB via inhalation. Neither duration nor
intensity of exposure is clearly known or stated in any of these
reports. In addition, it cannot be stated with certainty that each of
these persons was exposed to only p-DCB.

     Perrin (1941) reported dizziness and asthenia (weakness) in a
60-year-old man exposed to p-DCB in the bathroom. Petit and Champeix
(1948) reported the same symptoms in a 19-year-old female who prepared
p-DCB crystals in a factory for 18 months. Cotter (1953) reported
intense headaches in a 36-year-old woman who used p-DCB as a moth killer
in her home; he also reported headaches, nausea, and vomiting in a 34-
year-old woman who demonstrated products containing p-DCB. Persistent
headaches were among the symptoms reported by Cotter (1953) in a 60-
year-old man and his wife whose home had been saturated with p-DCB
mothball vapor for 3 or 4 months. The man also complained of numbness,
clumsiness, and a burning sensation in the legs. The man and woman both
died within a year of the initial exposure,  apparently as a result of
liver failure. It is important to note, however, that a definitive
association between the observed symptoms and results reported in these
case studies and p-DCB exposure has not been established.

     Inhalation, animal.  There are no acceptable data on CNS effects in
animals exposed to p-DCB by inhalation.

     Oral, human.  CNS effects in humans exposed to p-DCB via ingestion
have been reported in two case studies. Frank and Cohen (1961) reported
the case of a 19-year-old woman who ingested four or five p-DCB pellets
(p-DCB content unknown) daily for about 2.5 years with minor dermal
effects (patchiness). Upon ceasing consumption, she suffered tremors and
unsteadiness that were considered to be psychological rather than
physiological effects of withdrawal.

     A 21-year-old woman reported by Campbell and Davidson (1970)
developed pica (a craving for unnatural substances) for p-DCB toilet
bowl deodorizer blocks, which she consumed at the rate of one or two per
week throughout pregnancy. Reported CNS effects included tiredness,
dizziness, and mild anorexia. These effects, however, were not different
from the common general symptoms observed during normal pregnancy.

-------
40   Section 4

     These studies are not considered to provide adequate evidence that
ingestion of p-DCB by humans can result in CNS effects.

     Oral, animal.  There are no available studies which provide
evidence of CNS effects in animals exposed to p-DCB via oral exposure.
     Dermal, human.  There are no available studies which investigate
the potential CNS effects in humans dermally exposed to p-DCB.
     Dermal, animal.  There are no available studies which investigate
the potential CNS effects in animals dermally exposed to p-DCB.

     General discussion.  CNS effects have been reported in a number of
case studies of persons exposed to p-DCB via inhalation at unknown
levels. Although these data provide suggestive evidence that p-DCB can
cause CNS effects in humans exposed via inhalation, the information is
not sufficient to establish a defined dose-response relationship.
     Currently, there are no adequate data in humans or animals to
indicate that ingestion of or dermal contact with p-DCB can result in
CNS effects.

4.3.2.3  Liver effects

     Overview.   Exposure to p-DCB has resulted in adverse hepatic
effects in both humans and test animals. Suggestive evidence of hepatic
effects in humans has been demonstrated only in case studies of persons
exposed via inhalation. In animals, these effects have been observed
following short-term, intermediate, and chronic exposure via oral  and
inhalation routes and have been manifested as degeneration or necrosis,
sometimes coincident with porphyria. Effects on liver enzymes have also
been reported.

     Inhalation, human.  Hepatic effects have also been reported in
humans following long-term exposure to p-DCB via inhalation and possibly
via unverified dermal exposure. Cotter (1953) reported the cases of a
60-year-old man and his wife exposed to mothball vapor that "saturated"
their home for 3 to 4 months. Both of them died within a year of the
initial exposure, and acute yellow atrophy of the liver was confirmed at
autopsy. Subacute yellow atrophy and cirrhosis of the liver was the
diagnosis for a 34-year-old woman who demonstrated p-DCB products  in a
department store; a 52-year-old man who used p-DCB in his job in a fur
storage plant was also diagnosed with subacute atrophy of the liver
(Cotter 1953).  Duration of exposure was not estimated for either of the
latter two persons. No estimates of the p-DCB exposure levels have been
provided in any of the studies, nor was it verified that p-DCB exposure
was the only factor associated with the observed effects. Therefore,
available human case studies currently provide only qualitative evidence
that the liver is a target organ for p-DCB in humans.
     Inhalation, animal.  Hollingsworth et al. (1956) reported liver
effects in inhalation studies with p-DCB at various levels and durations
of exposure in rats and guinea pigs. Inhalation of p-DCB at levels of
158 ppm for 5 to 7 months by rats and guinea pigs for 7 h/day,
5 days/week caused cloudiness, swelling, and granular degeneration of
the liver, slight increases in the weights of the liver and kidneys, and
some interstitial edema and pulmonary congestion. Rats exposed to

-------
                                                 Toxicologies! Data   k\.

 341 ppm for 6 months showed evidence of a slight increase in the weight
 of the liver and kidneys. Guinea pigs exposed  to 341 ppm for 6 months
 exhibited hepatic changes including slight cirrhosis, focal necrosis,
 cloudiness, swelling, and fatty degeneration.  Based on various clinical
 and toxicological parameters,  the authors found exposure to p-DCB at
 96 ppm for 6 to 7 months to be without adverse effects in rats, guinea
 pigs, mice, rabbits, or one monkey. It should be noted, however, that
 this study was conducted with  several inconsistent variables including
 the species of animal at each  dose level, the number of animals per
 species exposed at each dose level, and the total number of exposures at
 each dose level. Therefore, the usefulness of  the resulting .data is
 limited, and the observed NOAEL of 96 ppm serves only as supportive
 evidence of the NOAEL of 75 ppm for hepatic effects reported by Riley et
 al. (1980) in a 76-week study  in rats as described below.

     In a long-term inhalation study, Riley et al. (1980, as summarized
 in Loeser and Litchfield 1983) exposed 76 to 79 male and female Alderly
 Park Wistar-derived rats to p-DCB at 0, 75, or 500 ppm for 5 h/day,
 5 days/week for 76 weeks with a subsequent 32- to 36-week observation
 period. SPF Alderly Park Swiss mice were also used in this study, but,
 due to the high incidence of respiratory infections in these animals,
 the results are not considered valid. In rats at the high exposure
 level, 500 ppm, there were reported increases in organ weights,
 including the liver, and slight increases in urinary protein and
 coproporphyrin output in the males. The NOAEL in rats was identified as
 75 ppm.

     Oral, human.   There are no available data on the hepatic effects of
p-DCB in humans exposed by ingestion.

     Oral, animal.  Hepatic effects have been reported in several
 studies in which p-DCB has been administered to test animals by gavage.
These effects have ranged from temporary effects on hepatic enzymes to
hepatic degeneration and necrosis.

     Short-term exposure.  p-DCB has been shown to produce disturbances
 in porphyrin metabolism after short-term exposure. Increased excretion
of porphyrins,  especially coproporphyrin and uroporphyrin, is considered
 to be an indication of liver damage.  Rimington and Ziegler (1963)
administered p-DCB to male albino rats at gradually increasing doses
until a dose level was reached (770 mg/kg/day for 5 days) which yielded
high porphyrin excretion but few fatalities. The first signs of
 intoxication were increases in urinary coproporphyrin and
porphobilinogen followed later by an increase in aminolevulinic acid
excretion. Mean peak values of urinary coproporphyrin increased to about
62 pg/day. 10-  to 15-fold when compared with controls. A nearly 100-fold
 increase in urinary uroporphyrin levels occurred, and porphobilinogen
 levels increased 200- to 530-fold when compared with controls. A 10-fold
 increase (to 437 Mg/day) in A-ALA levels was also observed, and
protoporphyrin levels increased 6-fold. Similar effects were not
observed when p-DCB was administered to rats at lower levels for a
 longer period of time by Carlson (1977), as discussed below.

     Miranda et al.  (1984) also reported that a single oral dose of
p-DCB at 800 rngAg administered to day-old white Leghorn chicks resulted

-------
42   Section 6

in significant increases  (P < 0.05) in the porphyrin concentrations of
both the liver and bile.

     Ariyoshi et al. (1975) investigated potential changes in certain
liver constituents, cytochrome P-450 levels, activities of some drug-
metabolizing enzymes (aminopyrine Af-demethylase and aniline
hydroxylase), and synthetase in rats treated with oral doses of p-DCB at
250 mg/kg/day for up to 3 days. Only A-ALA synthetase activity was
increased significantly by treatment with p-DCB (42%). The cytochrome
P-450 content did not change; however, the microsomal protein content of
liver preparations was increased.

     Carlson and Tardiff  (1976) studied the effects of several
halogenated benzenes, including p-DCB, on hepatic enzyme activities in
adult male rats. Groups of six rats were given p-DCB in corn oil by
gavage for 14 days. Significant decreases (P < 0.05) in hexobarbital
sleeping time were observed 24 h after cessation of a 14-day treatment
period at 650 mg/kg/day.  In addition,  when p-DCB was administered for 14
days at 20 or 40 mg/kg/day, increases were observed in hepatic
microsomal xenobiotic metabolism systems, including levels of glucuronyl
transferase and benzpyrene hydroxylase and EPN (0-ethyl 0-p-nitrophenyl
phenylphosphorothionate)  detoxification to p-nitrophenol.  At
10 mg/kg/day, other parameters measured, including cytochrome-c
reductase level, cytochrome P-450 content, and azoreductase levels, were
not affected. When these  authors conducted a 90-day study at the same
dosage levels, significant increases were seen in EPN detoxification,
benzpyrene hydroxylase, and azoreductase levels. The former two were
still elevated at 30 days after the cessation of administration of the
compound. Most increases were noted at 20 mg/kg/day and above, as in the
14-day studies; however,  azoreductase levels were elevated even at
10 mgAg/day.

     Intermediate exposure.  With administration of p-DCB to rats (two
per group) at 500 mg/kg/day by gavage for 4 weeks, marked hepatic
effects including cloudiness, swelling,  and centrilobular necrosis were
observed (Hollingsworth et al. 1956).  No adverse effects were observed
at 10 or 10Q mg/kg/day. The interpretation of this study is limited by
the size of the test groups and the fact that the use of controls was
not indicated.

     The NTP (1987) conducted two 13-week pilot studies in which F344/N
rats and B6C3F1 mice were dosed with p-DCB by gavage. In the first
study, rats were dosed at 300, 600, 900, 1200, or 1500 mg/kg/day, and
histologic changes were seen in the kidneys of male rats at all dose
levels. Based on these observations, doses in the second study were
lowered Co 37.5, 75, 150, 300, or 600 mg/kg/day.

     In the first study,  doses of 1200 or 1500 mg/kg/day produced
degeneration and necrosis of hepatocytes, hypoplasia of the bone marrow,
lymphoid depletion of the spleen and thymus, and epithelial necrosis of
the nasal turbinates in male and female rats. Liver weight to brain
weight ratios were increased at dosage levels of 900 mg/kg/day or more
for both male and female  rats. Serum cholesterol levels were increased
by doses of 600 mg/kg/day or more in male rats and 900 mg/kg/day or more
in female rats. Serum triglycerides and protein levels were reduced by
doses of 300 mg/kg/day or more in male rats. Urinary porphyrins were

-------
                                                 lexicological Data   43

increased in male rats administered 1200 or 1500 mg/kg/day and in female
rats receiving 1200 mg/kg/day. However, these increases were modest and
considered by the authors to indicate mild porphyrinuria rather than
hepatic porphyria. Liver porphyrins were not increased at any dose.
     In the second 13-week study, hepatic effects were not reported in
rats. Based on renal effects in the second study and all observed
effects in the first, a NOAEL of 150 mg/kg/day was identified for rats

     In the first NTP (1987) 13-week study in mice, p-DCB dose levels
were 600, 900, 1000, 1500, or 1800 mg/kg/day. Hepatocellular
degeneration was observed in both sexes at all doses, and the liver
weight to brain weight ratio was increased at doses of 900 mg/kg/day or
more. Serum cholesterol levels were increased in male mice at doses of
900 mg/kg/day or more, whereas serum protein and triglycerides were
increased at doses of 1500 mg/kg/day or more. These clinical chemistry
changes were thought by the authors to reflect the hepatic effects of
this compound. Hepatic porphyria was not found in mice at any dose in
this study.

     Because renal effects were seen in rats in all dose groups in the
first 13-week study, a second 13-week study was conducted at doses of
85, 169, 338,  675, or 900 mg/kg/day. Hepatocellular cytomegaly was
observed in male and female mice at doses of 675 and 900 mg/kg/day.
Based on the results of both 13-week studies, a NOAEL of 338 mg/kg/day
for hepatotoxicity was identified for mice. The lowest level at which
hepatic effects were observed (LOAEL) was 600 mgAg/day in the first
study.
                                                                 f
     Hollingsworth et al. (1956) reported liver toxicity in oral studies
with p-DCB.  Female rats (ten/group) were dosed with p-DCB at 0, 18.8,
188, or 376 mg/kg/day by gavage, 5 days per week for about 6 months. The
two higher doses resulted in an increase in the weights of the liver and
kidneys. At 376 mg/kg/day, decreased splenic weight and slight cirrhosis
and focal necrosis of the liver were also observed, as well as
cloudiness and swelling of the renal tubular epithelium. No adverse
effects were seen with the low dose. Therefore, 18.8 mg/kg/day was
identified as the NOAEL for the study, and 188 mg/kg/day was identified
as the LOAEL.  This phase of the Hollingsworth et al. (1956) study, as
compared with other phases described elsewhere in this profile, was
apparently controlled and reported.
     Data on the intermediate-term oral administration of p-DCB to
rabbits are also presented by Hollingsworth et al. (1956). However, the
level of administration was inconsistent and unclear (e.g., 263 doses of
500 mg/kg/day for 367 days to seven rabbits and "as many as" 92 doses of
1000 mg/kg in 219 days to five rabbits). The reported observations of
hepatic cloudy swelling and focal caseous necrosis are useful only as
supportive qualitative evidence of liver effects reported in other
studies.

     Carlson (1977) investigated the ability of p-DCB to induce
porphyria in rats when orally administered at 50 to 200 mg/kg/day for
30 to 120 days. Results indicated that p-DCB had little potential for
causing porphyria at these doses in female rats. After 30 and 60 days,
dose-related liver weight increases were observed, but even after

-------
44   Section 4

120 days, only slight increases in liver porphyrins occurred. Urinary
excretion of A-ALA, porphobilinogen,  or porphyrins was not increased
over control levels. Rimington and Ziegler (1963) had shown up to
100-fold increases in these parameters when p-DCB had been administered
at a higher level for a shorter period of time (i.e., 770 mg/kg/day for
5 days).
     Chronic exposure.  In the only study of lifetime oral exposure to
p-DCB, hepatic effects were seen in mice but not in rats (NTP 1987). In
this 2-year bioassay of p-DCB, male F344/N rats were dosed by gavage at
150 or 300 mg/kg/day and females at 300 or 600 mg/kg/day. Hepatic
effects were not observed in rats.
     In the same study,  male and female B6C3F1 mice were dosed by gavage
with p-DCB at 300 or 600 mg/kg/day. p-DCB increased the incidences of
nonneoplastic liver lesions in male and female mice, including
alteration in cell size (cytomegaly and karyomegaly), hepatocellular
degeneration, and individual cell necrosis.  Therefore, the LOAEL is
identified as 300 mg/kg/day.
     Dermal, human.  There are no available studies of the
hepatotoxicity of p-DCB in dermally exposed humans.
     Dermal, animal.  There are no available studies of the
hepatotoxicity of p-DCB in dermally exposed animals.
     General discussion.  Most of the available data on p-DCB toxicity
indicate that the liver is a major target organ for this compound. In
case studies in humans,  cirrhosis and subacute or acute yellow atrophy
of the liver have been diagnosed in individuals exposed to p-DCB via
inhalation at unknown levels and for uncertain durations. Data from
animal studies also demonstrated that p-DCB administered via inhalation
is hepatotoxic, with increased liver weights and increased porphyrin
excretion reported in one study and more severe changes, including
cirrhosis,  focal necrosis, and fatty degeneration, in another.
     In oral studies in animals, a broad spectrum of hepatic effects has
been reported, ranging from reversible effects on liver enzymes when low
levels were administered for brief durations (i.e., 20 mg/kg/day for
14 days) to hepatocellular degeneration and necrosis with chronic
administration of high levels (i.e.,  300 mg/kg/day for 2 years).
Porphyria has also been demonstrated with oral administration of p-DCB
in animal studies.

4.3.2.4  Renal effects

     Overview.  Adverse renal effects have generally been observed in
the same subchronic and chronic studies in which hepatotoxicity has
occurred. Effects have ranged in severity from increased kidney weights
to degeneration, mineralization, and hyperplasia of renal tissue.

     Inhalation, human.   There are no data available on the renal
effects resulting from human exposure to p-DCB via inhalation.

     Inhalation, animal.  In a long-term inhalation study, Riley et al.
(1980, as summarized in Loeser and Litchfield 1983) exposed male and
female Alderly Park Vistar-derived rats to p-DCB at 0, 75, or 500 ppm
for 5 h/day, 5 days/week for 76 weeks. SPF Alderly Park Swiss mice were

-------
                                                 ToxicoLog Leal Daca   45

 also exposed  to p-DCB,  but,  due  to  the high  incidence of  respiratory
 infections  in these  animals,  the  results  are  not considered valid. In
 rats at  the high exposure  level,  there were  increases in  organ weights,
 including the kidneys.  Based on  these and other (hepatic) effects, the
 NOAEL  in rats was  identified as  75  ppm.

     Hollingsworth et al.  (1956)  reported that inhalation of p-DCB at
 levels of 158 ppm  for 5  to 7  months by rats and guinea pigs over
 7 h/day, 5 days/week caused  a slight increase in the weight of the
 kidneys. Rats exposed to 341  ppm  for 6 months showed evidence of a
 slight increase in the weight of  the kidneys. Based on various clinical
 and toxicological parameters, the authors  found exposure  to p-DCB at
 96 ppm for 6  to 7 months to be without adverse effects in rats, guinea
 pigs, mice, rabbits, or one monkey. The limitations on the
 interpretation of these data  have been discussed, but the observed NOAEL
 of 96 ppm is  supportive of the NOAEL of 75 ppm for renal  effects
 reported by Riley et al. (1980) in  a 76-week study in rats previously
 described.

     Oral,  human.  There are  no data on the renal effects resulting from
 human ingestion of p-DCB.

     Oral,  animal.   In two NTP (1987) 13-week pilot studies, F344/N rats
 and B6C3F1 mice were dosed with p-DCB by gavage, as discussed under
 hepatic effects. In  the first study, rats were dosed with 300 to 1500
 mgAg/day.  Because histologic changes were observed in the kidneys of
 male rats at  all doses, a second 13-week study was performed at doses of
 38, 75, 150,  300, or 600 mg/kg/day. Renal  tubular cell degeneration was
 observed in male rats receiving 300 mg/kg/day or more in  the first
 study,  but only slight changes were seen at 300 mg/kg/day in the second
 study.  The kidney weight to brain weight ratio was increased in male
 rats receiving doses of 600 mg/kg/day or more. The blood urea nitrogen
 level was increased  slightly  in male rats dosed with 900 mg/kg/day or
 more.  Based on the combined results of both studies, a NOAEL of 150
 mg/kg/day was  identified in rats.

     In the two NTP  (1987) 13-week  studies in mice, dose  levels were 600
 to 1000 mg/kg/day and 80 to 900 mg/kg/day, respectively.  Renal effects
 in mice were not observed in  either study.

     Hollingsworth et al. (1956) observed renal toxicity  in oral
 studies with p-DCB.  In rats,   doses  of 188 or 376 mg/kg/day by gavage,
 5 days/week for about 6 months, increased the weights of  the kidneys. At
 376 mg/kg/day, cloudiness and swelling were observed in the renal
 tubular epithelium. No adverse effects were seen at the 18.8-mg/kg/day
 dose.

     Charbonneau et  al. (1987b) demonstrated that p-DCB administered by
 gavage to F-344 rats at 7 daily doses of 120 or 300 mg/kg/day
 significantly  increased (P <  0.05)  the level of protein droplet
 formation in  the kidneys of males but not in females. Administration of
 a single dose of ^C-p-DCB by gavage at 500 mg/kg gave similar results.
An analysis of the renal tissue of  animals administered radiolabeled p-
 DCB indicated  that it was reversibly associated with the protein a-2p-
 globulin. Bombard and Luckhaus (undated) designed a study to correspond
 to the experimental conditions of the 13-week NTP study with Fischer 344

-------
 46   Section 4

 rats.  p-DCB was  administered by gavage  in  corn oil at  0,  75,  150,  300,
 or 600 mg/kg/day for 13  weeks.  By 4  weeks, pronounced  cortico-medullary
 changes in the proximal  convoluted tubules of the nephrons  were  evident
 in most males at dosage  levels  of 150 mg/kg/day and higher. Cortical
 tubuli showed hyaline droplets  in the epithelia and large hyaline
 droplets and sporadic desquamated epithelia in the lumina.  At  13 weeks,
 one male at 75 rag/kg/day and all males  at  150 mg/kg/day and above  were
 observed to have an increased incidence of these changes. The  female
 animals showed no comparable findings.

      Renal effects have  also been observed in the only available study
 of chronic oral  exposure to  p-DCB. In the NTP (1987) bioassay, described
 above,  male rats exposed to  p-DCB at 150 and 300 mg/kg/day  for 2 years
 exhibited nephropathy, epithelial hyperplasia of the renal  pelvis,
 mineralization of the collecting tubules in the renal  medulla, and focal
 hyperplasia of the renal tubular epithelium. There were increased
 incidences of nephropathy in female  rats dosed with p-DCB at 300 and 600
 mg/kg/day (64 and 84%, respectively), as compared with vehicle controls
 (43%).  Therefore,  the LOAEL  for  renal effects in rats  was 150 mg/kg/day,
 the  lowest level tested  in this  phase of the study.

     p-DCB at 300 and 600 mg/kg/day  in this study also increased the
 incidence of nephropathy in  male mice and renal tubular degeneration in
 female  mice.  Therefore,  300  mg/kg/day was the LOAEL for renal effects in
 mice.

     Dermal,  human.   There are no  available data on the renal toxicity
 of p-DCB  in dermally  exposed humans.

     Dermal,  animal.  There  are  no available data on the renal toxicity
 of p-DCB  in dermally  exposed animals.

     General  discussion.   Evidence that p-DCB administration can result
 in renal  damage  comes only from  intermediate and chronic animal studies.
 In inhalation studies, increased renal weight has been reported.  In oral
 studies,  adverse  effects  ranged  from increased renal weight to
 degeneration  and mineralization  of the collecting tubules and focal
 hyperplasia of the  renal  tubular epithelium.

 4.3.3  Developmental  Toxicity

 4.3.3.1   Overview

     Limited  data suggested  that prenatal exposure to p-DCB results in
 developmental  toxicity at  maternally toxic levels.

 4.3.3.2   Inhalation

     Human.  There are no  available data on the developmental toxicity
 of p-DCB  in humans exposed to p-DCB via inhalation.

     Animal.  Hodge et al. (1977, as summarized in Loeser and Litchfield
 1983) conducted a teratogenicity study of p-DCB using pregnant SPF rats
exposed to p-DCB by inhalation at 0,  75. 200,  or 500 ppm for 6 h/day on
days 6 to  15 of pregnancy. These exposures did not result in
developmentally toxic effects in the offspring.  Therefore, 500 ppm is
 identified as  the NOAEL for  these effects.  (The terms NOEL and LOEL are

-------
                                                 lexicological Data   ^7

more commonly used  in evaluating developmental  toxicity, since all
effects, whether adverse or not, are considered important. In this
document, however,  the terms NOAEL and LOAEL are used instead).

     Hayes et al. (1985) similarly exposed pregnant New Zealand white
rabbits (28 or 30 per group) to p-DCB by inhalation at 100, 300, or 800
ppm for 6 h/day on  days 6 to 18 of gestation. The authors reported a
significant increase (P < 0.05) in the incidence of retroesophageal
right subclavian artery at 800 ppm. They concluded that it was a minor
variation in the circulatory system (seen in 2% of control litters in
their laboratory) and did not constitute a teratogenic response.
However, this effect was reported as an observed structural anomaly in
the development of  these fetuses; therefore, 800 ppm is the LOAEL for
this study. Slight maternal toxicity was also observed at 800 ppm as
indicated by significantly decreased body weight gain during the first 3
days of exposure. Therefore, 300 ppm is the NOAEL for maternal and
developmental toxicity.

4.3.3.3  Oral

     Human.  There are no available data on the developmental toxicity
of p-DCB in orally exposed humans.

     Animal.   In the only oral study identified, Giavini et al. (1986)
administered p-DCB to pregnant rats (13 to 17 per group) by gavage on
days 6 through 15 of gestation at 0, 250, 500,  750, or 1000 mg/kg/day.
At doses of 500 mg/kg and above, there was a dose-related significant
increase in the incidence of an extra rib in the fetuses (17, 29,  and
31% as compared to 6% of controls and 9% of 250 mg/kg/day dosage group;
P < 0.05,  P < 0.01,  and P < 0.01, respectively). A reduction in fetal
weights was observed at the 1000-mg/kg/day dose level, and maternal
weight gain was retarded at levels of 500 mg/kg/day and above.
Therefore,  250 mg/kg/day can be considered a NOAEL for maternal and
developmental toxicity, and 1000 mg/kg/day was  identified as the NOAEL
for fetotoxicity. (The reduction in fetal weight gain at 1000 mg/kg/day
was not considered to be a fetotoxic effect since it was associated with
a decrease in maternal weight gain at the same  dosage level.)

4.3.3.4  Dermal

     Human.  There are no available data on the developmental toxicity
of p-DCB in dennally exposed humans.

     Animal.   There are no available data on the developmental toxicity
of p-DCB in dermally exposed animals.

4.3.3.5  Discussion

     The available studies are limited but suggest that p-DCB is
developmentally toxic at levels that result in  maternal toxicity.  From
these studies it cannot be determined whether the observed developmental
toxicity is a result of maternal toxicity.

-------
48   Section 4

4.3.4  Reproductive Toxicity

4.3.4.1  Overview
     There are no appropriate data available to assess the potential
reproductive toxicity of p-DCB.

4.3.4.2  Inhalation

     Human.  There are no available data on the reproductive toxicity of
p-DCB in humans exposed via inhalation.

     Animal.  No reduction in reproductive performance (as measured by
the percentage of males successfully impregnating females) was observed
in an inhalation study by Anderson and Hodge (1976, as described by
Loeser and Litchfield 1983) in which male CD-I mice were exposed to
p-DCB at 75, 225, or 450 ppm for 6 h/day for 5 days before being mated
with virgin females. Studies to assess effects on male reproductive
performance, however, are usually conducted for the entire period of
spermatogenesis (about 9 weeks). Therefore, the conditions of this study
are not considered adequate to assess reproductive toxicity.

4.3.4.3  Oral

     Human.  There are no available data on the reproductive toxicity of
p-DCB in orally exposed humans.

     Animal.  There are no available data on the reproductive toxicity
of p-DCB in orally exposed animals.

4.3.4.4  Dermal

     Human.  There are no available data on the reproductive toxicity of
p-DCB in dermally exposed humans.
     Animal.  There are no available data on the reproductive toxicity
of p-DCB in dermally exposed animals.

4.3.4.5  Discussion

     Only one study, Anderson and Hodge (1976), investigates the
potential reproductive toxicity of p-DCB. As described above, only male
mice were exposed, the duration of exposure was only 5 days, and only
the inhalation route was tested. No conclusions can be drawn as to the
potential of p-DCB to induce reproductive toxicity based on the limited
database.

4.3.5  GenotoxicIty

4.3.5.1  Overview

     p-DCB is not mutagenic in most microbial and animal assay systems.
As indicated below, however, p-DCB is reported to be mutagenic in
plants. The results in microbial and mammalian systems are generally
considered to be more applicable to humans.

-------
                                                 ToxicoLogical Data   i9

4.3.5.2  Mlcrobial systems

     p-DCB was not mutagenic when  tested with cultures of histidine-
requiring mutants of Salmonella  cyphimuriua, in the Escherichia coli WP2
system, or in the Saccharomyces  cecevisiae C3 test for chromosomal
aberrations in yeast (NTP 1987,  Haworth et al. 1983, Anderson et al
1972, Anderson 1976, Simmon et al.  1979). However, p-DCB increased the
frequency of back mutation of the  methionine-requiring locus in the
fungus Aspergillus nidulans (Prasad and Pramer 1968, Prasad 1970).

4.3.5.3  Higher plants

     p-DCB has induced abnormal  mitotic division in several species of
higher plants. Observed effects  include shortening and thickening of
chromosomes, precocious separation of chromatids,  tetraploid cells,
binucleated cells, chromosomal breaks, and chromosomal bridges
(c-mitosis) (Sharma and Battacharya 1956, Sharma and Sarkar 1957,
Srivastava 1966, Gupta 1972).

4.3.5.4  Mammalian systems

     Unscheduled DNA synthesis was  nqt induced by p-DCB in cultures of
human lymphocytes (Perocco et al.  1983) or in HeLa cells (Institute di
Ricerche Biomediche (1986a). Instituto di Ricerche Biomediche (1987)
reported that p-DCB at concentrations up to 100 jig/mL, in the presence
or absence of metabolic activation, did not increase the incidence of
chromosomal aberrations in cultured human lymphocytes.

     Steinmetz and Spanggord (1987a,b) reported that gavage
administration of p-DCB at single  doses of 300 to 1000 mg/kg to B6C3F1
mice and Fischer 344 rats did not  result in unscheduled DNA synthesis in
the mouse hepatocytes or the renal  tissue of the rats. However, p-DCB ac
the highest level did induce an  increase in DNA replication (S-phase of
cell division) in the renal tissue  of the male rats and in the
hepatocytes of the male mice. Based on a comparison with historical
controls,  the authors concluded  that levels of DNA replication were also
significantly elevated in the livers of female mice.

     p-DCB did not induce chromosomal aberrations and sister-chromatid
exchange In Chinese hamster ovary cells at levels as high as ISO pg/mL
in the absence or presence of metabolic activation (S-9 from liver of
Aroclor 1254-induced male Sprague-Dawley rats) (NTP 1987).  Negative
results using this test were also  reported by Instituto di Ricerche
Biomediche (1986b) using p-DCB at  levels up to 100 jig/mL without
metabolic activation or up to 200 pg/mL with metabolic activation. p-DCB
did not increase the mutation frequency of L5178Y/TK+/' mouse lymphoma
cells with or without metabolic  activation (NTP 1987).

     Cytogenetic studies have been  conducted In rat bone marrow cells
following inhalation exposure of rats to p-DCB by Anderson and
Richardson (1976, as reported in Loeser and Litchfield 1983). Three
series of exposures were carried out: (1) one exposure at 299 or 682 ppm
for 2 h;  (2) multiple exposures  at  75 or 500 ppm,  5 h/day for 5 days;
and (3) multiple exposures to 75 or 500 ppm, 5 h/day, 5 days/week for
3 months.  Benzene and vinyl chloride were used as the positive controls,
negative controls breathed fresh air alone. Bone marrow cells from both

-------
SO   Section 4

femurs were examined  for chromosome or chromatid gaps, chromatic! breaks,
fragments, or other complex abnormalities. In all three experiments,
exposure  to p-DCB failed to induce any statistically significant effects
indicative of chromosomal damage when compared with the negative
controls.
     Cytogenetic effects were not found in bone marrow cells from male
and female B6C3F1 mice treated with p-OCB at levels up to 1800 mg/kg/day
in a 13-week study by NTP (1987). No increase in micronucleated cells
occurred  even at levels that were extremely toxic to the test animals
and resulted in liver toxicity and decreased survival. The NTP (1987)
study noted that the observed carcinogenic activity of this compound
cannot be adequately predicted on the basis of available genotoxicity
data, and that p-DCB may act as a tumor promoter rather than a mutagen.
     Herbold (1986) also reported no evidence of a clastogenic effect in
mouse bone marrow erythroblasts after a single oral administration of
p-DCB at  2500 mg/kg.

     Herbold (1986b) similarly found no evidence of clastogenic effects
in mouse bone erythroblasts after a single oral administration of 2,5-
dichlorophenol (the major metabolite of p-DCB) at 1500 mg/kg. Litton
Bionetics (1986) reported that 2,5-dichlorophenol with or without
metabolic activation did not induce an increase in mutagenic response in
the Chinese hamster ovary HGPRT forward mutation assay. This compound
was also  inactive in the Balb/3T3 in vitro transformation assay (Litton
Bionetics 1985). This profile has not attempted to present a full
evaluation of the mutagenic potential or the potential of any other end
points of the toxicity of 2,5-dichlorophenol.

4.3.5.5  Discussion

     The currently available data indicate that p-DCB is not mutagenic
in microbial or mammalian systems. The results of studies of higher
plants are of questionable significance in an evaluation of the health
effects of p-DCB in laboratory animals and humans. As mentioned above,
positive results in carcinogenicity testing of p-DCB combined with
negative results in mutagenicity testing suggest that p-DCB may act as a
tumor promoter rather than as an initiator in the carcinogenic process.
p-DCB has not been tested for its tumor promotion potential.

4.3.6  CareInogenlc1ty

4.3.6.1  Overview

     p-DCB has been shown to be carcinogenic in animal studies when
orally administered, but not following long-term inhalation exposure. In
the NTP (1987) 2-year carcinogenesis bioassay of p-DCB. there was clear
evidence of carcinogenicity in male rats and in mice of both sexes. No
human data are available regarding the carcinogenicity of p-DCB.

4.3.6.2   Inhalation

     Human.  There are no available data on the potential
carcinogenicity of p-DCB in humans exposed via inhalation.

-------
                                                  lexicological  Data   51

      Animal.   No evidence of carcinogenicity was observed in a  long-term
 inhalation study by Riley et al.  (1980,  as described by Loeser  and
 Litchfield 1983) using male and female SPF Alderly Park Wistar-derived
 rats.  SPS  Alderly Park Swiss mice were also used in this study,  but,  due
 to  the high incidence  of infections  in these animals,  the results  are
 not considered valid.  Rats were exposed to p-DCB at 0,  75,  or 500  ppm
 for 5  h/day,  5 days/week for 76 weeks.  As  mentioned in Sect.  4.2,
 Hawkins et al.  (1980)  has calculated that  tissue levels of p-DCB are
 similar when  repeated  oral doses  of  250  mg/kg/day or repeated inhalation
 doses  of 1000 ppm for  3 h/day are administered.  Based on the  reported
 lack of extensive organ toxicity,  the conditions of the Riley et al.
 (1980)  study  may have  resulted in considerably  lower tissue-levels  of
 p-DCB  than those occurring in animals in the NTP (1987)  gavage  study
 (150 and 300  mg/kg/day in male rats  and  300 and 600 mg/kg/day in female
 rats and mice  of both  sexes).  In  addition,  the  MTD was  not  achieved in
 the Riley  et  al.  (1980)  study.  Considering the  presumed lower effective
 dose in the rat-inhalation study,  as well  as the less-than-lifetime
 duration of dosing,  the  Riley et  al.  (1980)  study is not viewed  as
 providing  conditions adequate for  testing  the potential carcinogenicity
 of  p-DCB via  the inhalation route.

 4.3.6.3  Oral

     Human.  There are no  available  data on the  potential
 carcinogenicity  of p-DCB in humans exposed by ingestion.

     Animal.  p-DCB was  carcinogenic  in  both rats  and mice exposed  to
 p-DCB for  two years  in  the  NTP (1987) carcinogenesis bioassay. p-DCB was
 administered in  corn oil by gavage to F344/N rats  and B6C3F1  mice  in
 groups of  50 animals per sex per dosage  group. Male  rats  received doses
 of  150 and 300 mg/kg/day;  female rats and  mice of  both  sexes  received
 doses of 300 and 600 mg/kg/day.

     p-DCB produced a dose-related increase  in  the  incidence  of  tubular
 cell adenocarcinomas of  the  kidney in male  rats  [controls: 1/50  (2%);
 low dose: 3/50 (6%); high dose: 7/50  (14%)]. These  increases  occurred
with a significant (P -  0.005) positive  trend, and the  incidence in the
high-dose group was significantly greater  than that  in  the vehicle
 controls by the  life table  test.  [These  malignant  tumors are  uncommon  in
male F344/N rats; they have  been diagnosed  in only 4/1098  (0.4%) of the
 corn oil gavage  controls in previous NTP studies.) One  tubular cell
adenoma was observed in  a high-dose male rat. There  were no tubular cell
 tumors in dosed  or vehicle  control female  rats.  There was a marginal
 increase in the  incidence of mononuclear cell leukemia  in dosed male
 rats compared with that  in vehicle controls  [5/50  (10%);  7/50 (14%);
 11/50 (22%)]. This was only  slightly higher  than the incidence in
historical controls from the  same laboratory. The  NTP concluded  that
p-DCB was carcinogenic in male rats but  not  in female rats.

     p-DCB increased the incidences of hepatocellular carcinomas in
high-dose male mice  [14/50  (28%); 11/49  (22.5%);  32/50  (64%); ? < 0.01.
 Fisher Exact Test] and high-dose female  mice [5/50  (10%); 5/48 (10.4%);
 19/50 (38%); P < 0.01,  Fisher  Exact Test] and of hepatocellular adenomas
 in both high- and low-dose male mice  [5/50  (10%);  13/49  (26.2%); 16/50
 (32%); P - 0.006] and in high-dose female mice  [10/50 (20%);  6/48

-------
52   Section 4

(12.5%); 21/50 (42%); P - 0.008). Female control mice in this bioassay
had a substantially higher incidence of liver tumors than did historical
controls. Hepatoblastomas (a form of hepatocellular carcinoma) were
observed in four high-dose male mice (? - 0.015),  along with other
hepatocellular carcinomas, but not in vehicle controls. This rare
malignant tumor had not previously occurred in 1091 male vehicle control
mice in NTP studies. An increase in thyroid gland follicular cell
hyperplasia was observed in dosed male mice [1/47 (2%); 4/48 (2.3%);
10/47 (21.3%)], and there was a marginal positive trend in the incidence
of follicular cell adenomas of the thyroid gland in female mice [0/48
(0%); 0/45 (0%); 3/46 (6.5%)]. Pheochromocytomas (tumors of ehromaffin
tissue of the adrenal medulla or sympathetic preganglia) (benign and
malignant, combined) of the adrenal gland occurred with a positive trend
in dosed male mice, and the incidence in the high-dose group was
significantly greater than in the vehicle controls [0/47 (0%); 2/48
(4.2%); 4/49 (8.2%); P - 0.040]. The incidence of adrenal gland
medullary hyperplasia in male mice was 2/47 (4.3%), 4/48 (8.3%), and
4/49 (8.2%). Focal hyperplasia of the adrenal gland capsule was also
observed in male mice at 11/47 (23.4%), 21/48 (43.8%), and 28/49
(57.1%). (Unless otherwise stated, all P values presented for mice refer
to results derived using the Cochran-Armitage Trend Test. The results of
various other statistical tests have also been presented by the
authors.)

4.3.6.4  Dermal
     Human.  There are no available data on the potential
carcinogenicity of p-DCB to dermally exposed humans.
     Animal.  There are no available data on the potential
carcinogenicity of p-DCB to dermally exposed test animals.

4.3.6.5  Discussion
     p-DCB was carcinogenic in male rats and in male and female mice
when administered orally. As mentioned previously, further analysis of
the results of the NTP (1987) bioassay has raised questions as to the
relevance of the observed renal  tumors in male rats and hepatic tumors
in mice in evaluating the potential carcinogenicity of p-DCB in humans.

     The observation that kidney tumors are induced in male (but not
female) rats in response to exposure to certain chemicals has been  the
subject of recent research. Toxicologists at the CUT have hypothesized
that the male rat kidney is susceptible to the induction of certain
tumors  because it contains the protein a-2/i-globulin, which has not been
found at significant levels in  female rats or in mice or humans
(Charbonneau et al. 1987a). They have demonstrated  that a-2ft-globulin  in
combination with certain components of petroleum enhances the formation
of hyaline droplets in the proximal convoluted tubules of male  rats.
They hypothesize that the resulting cellular damage and cell
proliferation results in enhanced  tumor formation via a mechanism  that
has not yet been elucidated.  These  investigators are currently
conducting studies  to determine  whether similar processes occur during
the  induction of renal tumors in male  rats exposed  to p-DCB  (personal
communication  from  J. Swenberg,  CUT  1987). Kanerva et al.  (1987)  have

-------
                                                 Toxicological Daca   53

also demonstrated that the same effects can be elicited in male rats
orally administered either decalin (decahydronaphthalene) or d-liaonene
[l-methyl-4-(l-methylethenyl) cyclohexene] . Craig (1986) presented data
to show that a-2/j-globulin-associated nephropathy in male rats has been
observed in response to a broad range of chemicals (approximately 16 at
the time of his presentation). Based on these considerations, it is not
clear to what extent the rat kidney tumor  results are relevant in
determining the cancer risk to humans from inhalation of p-DCB.

     There has also been consideration of  the interpretation of the
finding of hepatocellular carcinomas and adenomas in mice in the NTP
(1987) study.  There was a high rate of these tumors in both male and
female control animals. Female control mice in this bioassay had a
substantially higher incidence of liver tumors than did historical
controls.  Because p-DCB has not been demonstrated to be mutagenic in any
of the microbial or mammalian systems tested, NTP (1987) has speculated
that it may act as a tumor promotor.

4.4  INTERACTIONS WITH OTHER CHEMICALS

     No studies have been identified which have investigated the effects
of p-DCB when administered with other chemicals.

-------
                                                                      55
               5.  MANUFACTURE, IMPORT, USE, AND DISPOSAL

5.1  OVERVIEW

     The three major production plants for p-DCB in the United
States are located in Illinois, West Virginia, and Delaware. The
major uses of p-DCB are as space deodorizers (primarily in bathrooms)
(20 million Ib), moth repellents (8 million Ib),  and an intermediate in
the production of polyphenylene sulfide (PPS) resins (22 million Ib).
These three uses accounted for over 90% of the p-DCB used in the
United States in recent years. Of the total U.S.  annual production of
75 million Ib, about one-third (or 25 million Ib) is exported.
Approximately 95% of the environmental releases of p-DCB occur during
its use, rather than during its manufacture or processing. Relatively
small amounts of p-DCB are disposed of in landfills from production
processes.

5.2  PRODUCTION

     p-DCB is produced as a by-product of monochlorobenzene
manufacturing in a process involving direct chlorination of benzene in
continuous or batch reactors. The products include mono- and
dichlorobenzenes and insignificant amounts of higher chlorinated
benzenes. The p-DCB fraction is separated by distillation.

     The batch chlorination process yields a higher proportion of
dichlorobenzenes than the continuous process and can be modified to
yield an even greater proportion of p-DCB (EPA 1981).  p-DCB can also be
produced by the Sandmeyer procedure from the appropriate chloroaniline
(HSDB 1987).

     p-DCBs can be manufactured in higher yields using a continuous
process by chlorinating benzene in the presence of aluminum chloride
(Lowenheim and Morgan 1975).

     The total 1984 production of about 75 million Ib was produced
mostly by three companies in Illinois, West Virginia,  and Delaware (ICF
1987).

5.3  IMPORT

     Since 1980 virtually no p-DCB has been imported into the United
States.   About one-third of the total production of p-DCB has been
exported in recent years (ICF 1987).

5.4  USE

     One major consumptive use of p-DCB is as a space deodorizer for
toilet bowls,  urinals, garbage cans, and diaper pails.  These products
are usually 100% p-DCB blocks, sometimes with added perfume. A second

-------
56   Section 5

Important use of p-DCB Is moth control, in the form of mothballs or moth
repellent blocks. The third important use is as a chemical intermediate
in the production of PPS resins. Together, these three uses accounted
for over 90% of the p-DCB used in the United States in 1984 (ICF 1987).
p-DCB is also used in pesticides, dye synthesis, abrasives, floor waxes
and finishes, and agricultural chemicals (HSDB 1987, EPA 1981).

5.5  DISPOSAL

     It is estimated that approximately 1,000,000 Ib,  or 1.5% of total
p-DCB production during 1978,  was released to the environment during
manufacture. Approximately 17,000 Ib, mainly in sludge from
fractionating towers (EPA 1981), was disposed of in landfills from
production processes in 1978.  About 640,000 Ib of p-DCB was discharged
to water.  Because p-DCB sublimes at room temperature (20°C),  some of
this waste may also reach the air.

-------
                                                                       57
                          6.   ENVIRONMENTAL FATE

 6. 1  OVERVIEW

      p-DCB enters the environment primarily as a result of releases
 during use.  Manufacturing accounts for only -1.5% of the environmental
 releases.  There are no natural sources of p-DCB.

      Relatively little information concerning the environmental  fate  of
 p-DCB is available.  p-DCB is expected to volatilize  at  a relatively
 rapid rate,  allowing atmospheric transport.  It has an estimated  half-
 life  of 9  h  or less for removal from surface water via  evaporation. The
 dichlorobenzenes are reported to be reactive toward  hydroxyl  radicals in
 air with a half-life of about 3 days,  but indirect evidence suggests
 that  p-DCB does not hydrolyze at a significant rate  under normal
 environmental  conditions.  Other estimates indicate p-DCB residence time
 in  the  atmosphere of more than 38 days.  Its  high  log octanol/water
 partition  coefficient suggests that adsorption to organic matter in
 aquatic systems and soil  is  probably an important environmental  fate
 process. Indirect evidence suggests that bioaccumulation,  may also be an
 important  fate  process. p-DCB appears  to be  initially resistant  to
 biodegradation  in soil; however,  it may be broken down  to some degree by
 pollutant-acclimatized microorganisms.  Sorption,  bioaccumulation, and
 volatilization  with  subsequent atmospheric oxidation are  likely  to be
 competing  processes,  with the dominant  fate  being determined by  local
 environmental conditions.  If volatilization  does  not occur, p-DCB is
 probably persistent  in the environment  (e.g.,  when in groundwater).

 6.2  RELEASES TO  THE  ENVIRONMENT

 6.2.1  Anthropogenic  Sources

     In 1984, almost  30 million Ib  of p-DCB  was used in  the United
 States  in  the form of space  deodorants  and moth repellents. All  of the
 20 million Ib used in space  deodorants  is assumed to  be released to the
 environment, with about 90%  released to  the  air and  the remainder to
 land or water. Another major  use  of p-DCB, moth control,  consumes 8 to
 12 million Ib annually, all  of which is  assumed to be released to the
 atmosphere. Minor uses of  p-DCB  and inadvertent sources contribute
 little to the environmental  releases of  the  compound (ICF 1987,   EPA
 1981). Taken together, the above  information indicates that little p-DCB
 enters the atmosphere  from hazardous waste disposal  sites. As summarized
 in Sect. 7, use of consumer products accounts  for  the greatest source of
p-DCB in the atmosphere.

-------
58   Section 6

6.2.2  Natural Sources
     There are no natural sources of p-DCB.

6.3  ENVIRONMENTAL FATE

6.3.1  Atmospheric Fate Processes

     Atmospheric p-DCB may theoretically be  degraded by chemical- or
sunlight-catalyzed reactions or may be adsorbed onto particles that
settle or are removed from the atmosphere by rain.  One measure of the
effectiveness of these factors in removing p-DCB from the air is the
atmospheric residence time. The atmospheric  residence time is the time
for removal of all of the chemical initially present in the atmosphere.
Singh et al. (1981. as cited in EPA 1985) reported that the atmospheric
residence time for p-DCB was 38.6 days. This estimate is based on a
daily average hydroxyl radical abundance of  106 mol-cm"^ in the boundary
layer of a polluted atmosphere, which is equivalent to a daytime (12 h)
hydroxyl radical abundance of 2 x 10" mol-cm"-* (typical of summer
months, but much higher than that expected during winter). These authors
also estimated a daily loss rate of 2.6% (assuming 12 h of sunlight per
day).
     Dichlorobenzenes in general were reported by Ware and Weast (1977,
as cited in EPA 1979) to be reactive with hydroxyl radicals in the air
with a half-life of approximately 3 days. The half-life is the time
required for removal of one-half of the initial concentration of the
chemical. An estimate of the corresponding atmospheric residence time
was not provided in the cited reference. Thus, the relationship or
consistency between this half-life estimate  and the residence time
estimate summarized above is not readily apparent.  These authors also
reported that p-DCB is resistant to auto-oxidation by ozone in air.
     Brunce et al. (1978) report that photolysis of chlorinated
biphenyls in iso-octane solution involves a  triplet reactive state and
that excimers do not appear to play a role in the photodegradation.
Their study did not include p-DCB.

6.3.2  Surface Vater/Groundvater Fate Processes
     The available data indicate that degradation of chlorobenzenes in
aquatic systems may be carried out by microbial communities in
wastewater treatment plants and in natural bodies of water (EPA 1985).
The degradation of p-DCB in aquatic systems  is believed to be highly
variable and dependent upon specific microbial (and possibly other)
conditions. This is evidenced by a monitoring study in the Rhine River
that indicated the degradation half-life ranged from 1.1 to 25 days
based on a limited number of individual measurements from different
sampling locations (EPA 1985).
     Ware and Weast (1977) indicate that p-DCB is resistant to auto-
oxidation by the peroxy radical (RO.•)  in water. Indirect evidence
suggests that p-DCB does not hydrolyze  at a significant rate under
normal environmental conditions. This  is believed to be due to the
extreme difficulty with which the substance  undergoes nucleophilic
substitution (EPA 1979).

-------
                                                 Envirorwencal Face   59

     The available  data  also  indicate  that p-DCB probably volatilizes
 from the water column  to the  atmosphere at a  relatively rapid rate. EPA
 (1979) used the data of  Garrison and Hill (1972) to calculate an
 approximate evaporative  half-life of less than  30 min for aerated
 solutions; the data for  unaerated conditions  correspond to a half-life
 of  less than 9 h.

     Although no specific  environment  sorption  studies were located, the
 log P value of 3.39 (Leo et al. 1971,  as cited  in EPA 1979) indicates
 that sorption processes  may be significant at p-DCB concentrations
 anticipated in natural waters. The adsorption of p-DCB to sedimentary
 organic material may compete  with volatilization processes to the
 atmosphere (EPA 1979).

 6.3.3  Soil Fate Processes

     Studies of the fate of the dichlorobenzenes in soil indicate that
 these compounds are somewhat  resistant to microbial degradation.
Alexander and Lustigman  (1966, as cited in EPA  1979) stated that the
presence of a chlorine atom on the benzene ring retarded the rate of
degradation;  therefore,  p-DCB, being more highly chlorinated than
chlorobenzene,  would presumably biodegrade no faster than chlorobenzene
under similar environmental conditions (EPA 1979). Ware and Weast
 (1977), Lu and Metcalf (1975), and Chiou et al. (1977. as cited in EPA
1979) present data  which show chlorobenzene to be resistant to
biodegradation,  unless microorganisms are already growing on another
hydrocarbon source  in the medium.

     Ballschmiter and Scholz  (1980, as cited in EPA 1985) investigated
the metabolism of p-DCB  by the soil microbe Pseudomonas and found that
this bacterium degraded  the compound to dichlorophenols and
dichloropyrocatechols.  Monsanto (1986) reported that p-DCB is moderately
to readily biodegradable under their assay conditions. Specifically,
primary degradation was  greater than 95% in a 24-h, semicontinuous
activated sludge test.  Using  the Thompson-Duthie-Sturm assay method,
p-DCB yields  a theoretical C02 evolution of approximately 58%. These
assay results probably are not representative of the biodegradation
levels likely Co occur under  typical soil conditions;  however, they do
represent fate  under enrichment conditions.

6.3.4  Biotic Fata  Processes

     The log octanol/vater partition coefficient (log p - 3.39,  Leo et
al.  1971,  as  cited  in EPA 1979) suggests that p-DCB has a high potential
for bioaccumulation in aquatic organisms.  The incorporation of chlorine
into an organic  molecule generally increases its lipophilic character
and bioaccumulation potential (Kopperman et al. 1976,  as cited in EPA
1979);  therefore,  p-DCB  should be more lipophilic than benzene or
chlorobenzene.

-------
                                                                       61
                     7.  POTENTIAL FOR HUMAN EXPOSURE

 7.1  OVERVIEW

      p-DCB has been detected in air, water, soil,  and foods.  Potential
 human exposure can result from inhalation or via ingestion of food or
 water.  Inhalation is likely to be the most significant route  of human
 exposure.  There are no data which indicate that ingestion of
 contaminated food is a significant exposure route.  Additionally,
 exposure to-substantial amounts of p-DCB from drinking contaminated
 water appears to be low for the general population.  However,  the
 potential  for substantial,  but localized,  human exposures as  the  result
 of  contaminated groundwater near hazardous waste sites has been shown.
      Human exposure to p-DCB is greatest in indoor  air where  higher
 exposure potentials exist to vapors from p-DCB products used  as
 deodorizers and moth repellents.  Various studies suggest that indoor  air
 concentrations  are  8 to 100 times greater than outdoor ambient levels.
 Specific measurements  of indoor air concentrations  for combined m-DCB
 and p-DCB  at several U.S.  locations provide estimated mean values
 ranging  from 5.5 to 56 /ig/m3 (approximately 0.0009  to 0.009 ppm),
 whereas  estimated mean values for outdoor concentrations were
 approximately an order of magnitude less (0.3  to 2.2  A»g/m3; 0.00005 to
 0.0004 ppm).  Median values  ranged from 0.53 to 4.2 A»g/m3 (0.0001  to
 0.0007 ppm)  for indoor air  and from 0.07 to 1.7 Mg/m" (0.00001 to  0 0003
 ppm)  for outdoor air.

      Populations at elevated risk for exposure to high concentrations of
 p-DCB include workers  involved in the manufacture and processing of the
 compounds  and those individuals who may be  transiently exposed to  high
 levels of  p-DCB in  moth repellents.  Other occupational groups with
 suspected  elevated  exposures to p-DCB include  janitors,  furriers,  and
 undertakers.  No populations  with  above-average sensitivities  to p-DCB
 have  been  identified.

 7.2   LEVELS MONITORED  OR ESTIMATED  IN THE ENVIRONMENT

 7.2.1  Levels in Air

      In discussing  atmospheric  levels of p-DCB,  it is  necessary to
 distinguish between indoor air  and  outdoor  (ambient)  air.  The
 distinction  is  important because  household  consumer products  can release
 large amounts of p-DCB  to the  indoor  air, and  indoor  air concentrations
 may be much higher  than outdoor concentrations.

     The most comprehensive  data  on  indoor  air  concentrations  of p-DCB
 in the United States comes from the  EPA  Total  Exposure Assessment
Methodology  (TEAM)  study (Wallace et  al. 1986).  Using  personal monitors.
combined concentrations of m-DCB  and  p-DCB  were  obtained for  persons  in

-------
62   Section 7

approximately 600 homes in New Jersey,  North Carolina,  North Dakota,  and
California. (The combined measure was assumed to be almost entirely p-
DCB.) The highest mean nighttime personal exposures (taken as an
indication of concentrations in homes)  were reported to be 49 to 56
pg/m3 (0.008 to 0.009 ppm) in the New Jersey study area,  with a maximum
measured value of 1500 /ig/m3 (0.25 ppm).  Mean personal  exposure
concentrations for the other three areas  ranged from 5.5 to 18 ^g/m3
(0.0009 to 0.003 ppm). By contrast, outdoor mean concentrations for the
four areas ranged from 0.3 to 2.2 pg/m3 (0.00005 to 0.0004 ppm). Median
concentrations from nighttime personal monitoring (indoor air) in the
four areas ranged from 0.53 to 4.2 Mg/m3  (0.0001 to 0.0007 ppm).
     Other studies also document p-DCB in the indoor air environment.
Scuderi (1986) measured the rate of increase of air concentrations of
p-DCB after p-DCB blocks were placed in public bathrooms or p-DCB
crystals were placed in a wardrobe. The concentration in one of the
bathrooms exceeded 200 /ig/m3 (0.03 ppm).  Lebret et al.  (1986), in a
study of 300 Dutch homes, reported that indoor p-DCB concentrations
reached 300 ^g/m3 (0.05 ppm), whereas outdoor maximum and median
concentrations were less than 0.6 /*g/m3 (0.0001 ppm).  Morita and Ohi
(1975) reported maximum outdoor p-DCB concentrations in Tokyo of 4 jjg/m3
(0.0007 ppm), whereas they observed that the p-DCB concentration in a
bedroom was more than 100 A»g/m3 (0.02 ppm).
     Additional outdoor measurements in the United States also showed
generally lower levels than reported for some indoor environments.
Bozzelli and Kebbekus (1982) reported a mean concentration of 1.7 /jg/m3
(0.0003 ppm) from 330 outdoor samples taken in varied locations in New
Jersey in 1978. Brodzinsky and Singh (1982, as cited in EPA 1985)
reported a mean p-DCB concentration of 1.6 pg/m3 (0.0003 ppm) for 24
localities. Wallace et al. (1986) found that m-DCB and p-DCB were
ubiquitous; that is, they were detected in 80 to 100% of the samples at
each TEAM site.
     The data presented above indicate that indoor air is a major source
of exposure to p-DCB. However, data needed to generate population
frequency distribution curves of exposure are limited.

7.2.2  Levels in Water
     p-DCB has been detected in surface water and groundwater and in
municipal and industrial discharges. The EPA Storage and Retrieval
(STORET) database system contains monitoring data on this chemical. The
EPA  (1975) presented the results of a survey of groundwater and surface
water at more than 1000 sites throughout New Jersey. Isomers of
dichlorobenzenes were found  in about 3% of all groundwater samples and
in 4% of the surface water samples.
     The TEAM study by Wallace et al. (1986) also reported on the
combined occurrence of m-DCB and p-DCB in drinking water  in selected
cities in New Jersey, North  Carolina, and North Dakota. These DCBs were
generally classified as "occasionally found," being detected  in 3% of
samples in New Jersey, 2% of samples in North Dakota, and 0% of samples
in North Carolina. Coniglio  et al.  (1980, as cited in EPA 198S) reported
that 13% of  the drinking water treatment plants in the United States
that use groundwater had drinking water samples that contained  p-DCB.

-------
                                        Potential  for Human Exposure   63

 The  reported concentration  range  for  all  dichlorobenzene  isomers was <1
 to 100 Mg/L. Oliver  and  Nlcol  (1982.  as cited  In  EPA 1985) sampled
 drinking water  in  three  cities  on Lake  Ontario. p-DCB concentrations
 ranged from 8 to 20  ng/L (mean  -  13 ng/L) .  The National Organics
 Monitoring Survey  (Symons et al.  1975,  as cited in  EPA 1981) found p-DCB
 in 2/111. 20/113.  and  29/110 samples  (representing  three  different
 sampling phases) of  drinking water analyzed during  the period March 1976
 to January 1977. The mean concentration ranged from 0.07  to 2.0 ng/L,
 and  the median  concentration was  <1 Mg/L  for all  samples  in all phases
 of the survey.  These data are generally consistent  with the results of
 an earlier EPA  study (EPA 1975, as cited  in EPA 1981), which indicated 1
 Mg/L as the highest  drinking water concentration  found for p-DCB.

     Chlorobenzenes  have also been detected in wastewaters from
 industrial processes and in influents and effluents at municipal sewage
 treatment plants.  Gaffney (1976,  as cited in EPA  1985) sampled water at
 four municipal  facilities which handled sewage and  wastewater from
 carpet mills. Average  dichlorobenzene (all  isomers) concentrations in
 the  incoming and outgoing water ranged  from 3 to  146 Mg/L and 0 to 268
 Mg/L, respectively.  The author concluded that the increases in effluent
 dichlorobenzene levels resulted from chlorination (e.g.,  by reaction of
 chlorine with precursor organics  in the water). Neptune (1980, as cited
 in EPA 1985) compiled  data collected during several industrial
 wastewater surveys conducted from September 1978  through October 1979.
 p-DCB concentrations greater than 10 Mg/L were detected in 88 of 3268
 samples (range - 10  to 410 Mg/L;  median - 79
     Significant localized contamination of drinking water may occur if
hazardous waste site leachate enters either surface water or groundwater
supplies. Several limited studies provide some information on this
exposure potential. Bramlett et al. (1986) performed priority pollutant
analyses of the leachate from 13 hazardous waste sites. Dichlorobenzenes
and monochlorobenzene accounted for 1.4% of the total mean mole fraction
of all organic chemicals detected in the leachates. The combined
chlorobenzenes were detected in the leachates from 6 of the 13 sites.

     Ghassemi eC al. (1984) compiled a database on the constituents in
hazardous waste site leachates from 30 sets of analytical data collected
from 11 different sices. Dichlorobenzene was detected in one of eight
leachate samples of "mixed industrial wastes" collected above liners and
in five of seven leachate samples of "mixed industrial wastes" collected
below or between liners. The highest average concentrations of
dichlorobenzene in these samples were reported to be less than
0.311 mg/L for the sample collected above liners and 0.67 mg/L for the
samples collected below or between liners.

     Wetzel et al. (1985) detected dichlorobenzene (detection limit of
0.001 mg/L) in one of seven groundwater wells in the vicinity of a waste
disposal site at Kelly Air Force Base, Texas.

     The potential for water contamination depends on the quantity of
DCB disposed of at a particular site.  The environmental fate data
presented earlier, however, indicate that p-DCB will be relatively
stable and persistent in water not exposed to air. Thus, the potential
exists for significant contamination of groundwater and/or surface water

-------
 64    Section  7

 at  some waste sites. However,  specific occurrences have not been
 documented  in the  literature  located for this report.

 7.2.3 Levels in Soil

      The  available  studies  of  soil contamination by chlorobenzenes have
 focused on  hexachlorobenzene with relatively few surveys including all
 chlorobenzenes. Elder et  al.  (1981, as cited in EPA 1985) detected all
 of  the chlorobenzenes in  the part-per-million range in the sediments of
 streams draining the Love Canal area of Niagara Falls, New York. Oliver
 and Nicol (1982, as cited in EPA 1985) detected all chlorobenzene
 isomers in  the sediments  of Lakes Superior, Huron, Erie, and Ontario.
 The most  contaminated lake  was Lake Ontario, with dichlorobenzene
 sediment  levels ranging from 11 to 94 ng/g.

      Wetzel et al.  (1985) detected dichlorobenzene in soil samples from
 three of  five boreholes in  and around a waste disposal site at Kelly Air
 Force Base, Texas. Concentrations ranged from 0.15 to 38.0 mg/L of soil.
 These data  further substantiate the potential for localized groundwater
 contamination in the vicinity of some waste sites.

 7.2.4 Levels in Food

      Dichlorobenzenes may be present in food as a result of
 contamination from uses of  the chemical.  Schmidt (1971, as cited in EPA
 1980) reported the contamination of pork as a result of the use of p-DCB
 in  pig stalls as an odor-control product.  Additionally, hens exposed to
 p-DCB concentrations of 20  to 38 mg/m3 produced contaminated eggs within
 3 days of exposure (Langner and Hillinger 1971,  as cited in EPA 1980).
 Morita et al. (1975, as cited in EPA 1980)  reported detectable levels of
 p-DCB in fish from Japanese coastal water;  a species of mackerel
 contained 0.05 mg/kg p-DCB. Oliver and Nicol (1982, as cited in EPA
 1985) detected all dichlorobenzene isomers  in trout from the Great
 Lakes.

      Limited data from 42 samples of human breast milk at five U.S.
 locations .detected DCB (all isomers) at concentrations ranging from
 0.04  to 68 ng/mL (Cone et al. 1983).

      A measured steady-state bioconcentration factor of 60 (which
 relates the concentration of a chemical in aquatic animals to the
 concentration in the water  they inhabit)  was determined for p-DCB, using
bluegills (EPA 1978a,b. as cited in EPA 1980).  When this factor is
 adjusted for the percent  lipid in bluegills compared with the weighted
 average percent lipids in the edible portion of fish and shellfish
 consumed in the United States, a weighted average bioconcentration
 factor of 37.5 is derived for p-DCB in aquatic organisms consumed in the
United States (EPA 1980).  For example,  a concentration of 1 mg/L in
water would result in a concentration of 37.5 ppm (wet weight) in the
 fish  edible portion.

 7.2.5  Resulting Exposure Levels

      The monitoring studies discussed in the previous sections indicate
 that p-DCB is present in  the environment,  and human exposure is more
 likely to result from the Inhalation of contaminated air (indoor and

-------
                                        Potential for Human Exposure   65

 outdoor)  than from the ingestion of contaminated water or food.  The
 compound  is fairly ubiquitous in environmental media at low levels,
 although  substantial human exposure may occur from contact with  certain
 media (e.g.,  indoor air,  localized contaminated water,  and air in some
 occupational  environments).  Evidence of widespread human exposure to
 p-DCB is  provided by data from EPA's National Human Adipose Tissue
 Survey (NHATS)  (EPA 1986c).  Results from the NHATS FY 1982 repository
 show that p-DCB was detected in all 46  composite samples of human
 adipose tissue  from all U.S.  regions. Wet tissue concentrations  ranged
 from 0.012 to 0.50 Mg/g-  No  summary conclusions were drawn regarding the
 human health  significance of these levels.

      Because  the  largest  environmental  releases of p-DCB are  to  the air,
 and  because volatilization is a major process for removing the substance
 from aquatic  media,  inhalation is  expected to be the major p-DCB
 exposure  route  for the general population.  In a recent  estimate  of human
 exposure  to p-DCB from the atmosphere by the EPA Office  of Drinking
 Water, the median concentration for outdoor urban/suburban environments
 of 0.28 A»g/m3 (0.00005 ppm) was used (EPA 1987a).  This  is  generally
 consistent with the  TEAM  study findings  in which the median outdoor air
 concentrations of p-DCB ranged from 0.07 to 1.7 Mg/m3  (0.00001 to  0.0003
 ppm)  (Wallace et  al.  1986). By contrast,  the TEAM study  reports  median
 indoor air concentrations  of  p-DCB in the range of 0.53  to 4.2 /ig/m3
 (0.0001 to 0.0007  ppm)  and mean concentrations in the range of 5.5 to 56
 Mg/m3  (0.0009 to  0.009  ppm)  (Wallace et  al.  1986).  When  making human
 exposure  estimates  for a  70-kg adult, a  breathing rate of  20 m3/day is
 typically  used.

     The most direct  route of exposure for  waterborne p-DCB is the
 ingestion  of drinking water.  The EPA (1981)  estimated human exposure to
 p-DCB via  contaminated water  based on National Organics  Monitoring
 Survey data. These estimates,  which assume  daily water consumption of
 2 L/day, are presented  in Table  7.1. A recent  exposure assessment of
 p-DCB by  the EPA Office of Drinking Water (EPA 1987a) estimated  that
 over 1.6 million persons  (0.8%  of  the population served  by  public water
 supplies)  were exposed  to p-DCB at drinking water concentrations at or
 above 0.5  Mg/L, but that no individuals  receive  drinking water exposures
 above 5 Mg/L.  Surface water (53%)  and groundwater (47%)  contribute
 nearly equally to these exposures.  These  estimates  indicate the
 potential   for exposure  to substantial amounts  of p-DCB from drinking
water appears to be low. However,  based  on  the  limited soil and  water
 data from hazardous waste sites, localized  contamination may result in
 substantially increased water  exposures  for some  populations.

     Although p-DCB contamination  of food occurs,  there  are no data
 indicating that this  is a significant route  of human exposure (EPA
 1981).

 7.3  OCCUPATIONAL EXPOSURES

     Levels of p-DCB may be much higher  in  certain workplace atmospheres
 than in ambient air. Ware and Weast (1977,  as cited  in EPA  1985)
reported that in workplaces associated with  the manufacture of p-DCB,
atmospheric concentrations of  the  chemical  averaged  204  mg/m3 (34 ppm)
 [range - 42 to 288 mg/m3 (7 to 48  ppm)]  near shoveling and  centrifuging

-------
66    Section 7
                     Table 7.1. Estimated hommn exposure to p-DCB



Maximum observed
Mean observed
Median concentration
Estimated
Concentration
(Mg/L)
2.0
0.07
<0.005
exposure
Exposure
(mg/day)
4 X 10~3
io-4
<10~5
                     Source: Adapted from EPA 1981.

-------
                                       Potential for Human Exposure   67

operations and  150 mg/m3  (25 ppm)  [range -  108  to 204 mg/m3 (18 co 34
ppm)] during pulverizing  and packing. Generally, however, levels
throughout the  manufacturing plant were detected at less than 48 mg/ra3
(8 ppm). Pagnatto and Walkley  (1965, as cited in EPA 1981) reported
concentrations  of 150 to  420 mg/m3 (25 to 70 ppm) in workplaces
associated with the manufacture of p-DCB. NIOSH (1980) reported that a
worker at a p-DCB drumming operation was exposed to -30 mg/m3 (5 ppra,
8-h TWA), and that some workers are exposed to  levels up to 220 mg/m3
(37 ppra). Measurements made by the Occupational Safety and Health
Administration  (OSHA 1986) between 1981 and 1986 show numerous instances
of workers exposed to greater than 66 mg/m3 (8 ppm).

7.4  POPULATIONS AT HIGH  RISK

7.4.1  Above-Average Exposure

     Subpopulations in the vicinity of production or industrial use
facilities may  be exposed to transitory higher levels of p-DCB due to
accidental releases, but  there are no data upon which to base an
estimate of this type of  exposure (EPA 1981).

     Workers involved in  the production of p-DCB may be exposed to
concentrations  significantly higher than those encountered by the
general population.  A NIOSH survey reported by Hull and Co.  (1980, as
cited in EPA 1981) projected that approximately one-half million workers
had nfull"-time (>4 h/week) or "parf-time (<4 h/week) exposure to p-
DCB.  Hull and Co.  estimated on the basis of their own survey that only
approximately 821 employees of manufacturers,  processors, formulators,
and resellers of p-DCB would be potentially exposed.

     High exposure levels may result from some consumer use of moth
repellents and  room deodorizers.  The indoor air measurements by Wallace
et al.  (1986) suggest that consumers who use p-DCB deodorizers and moth
repellents will have higher exposures than persons who are not regularly
exposed to these products. Although the TEAM study provided no
information on  the household use of products containing p-DCB, studies
now under way are expected to confirm that higher indoor air
concentrations of p-DCB are related to household uses of products
containing p-DCB.

7.4.2  Above-Average Sensitivity

     No Information is available on populations with above-average
sensitivity to p-DCB.

-------
                                                                       69
                          8.  ANALYTICAL METHODS

      Gas chromatography (GC) is the most common analytical method for
 detecting and measuring p-DCB in environmental and biological samples.
 High-performance liquid chromatography may also be used to measure p-DCB
 in environmental samples (Bush et al.  1984).

      Samples may be prepared by several methods,  depending on the matrix
 being sampled.  The sample preparation procedures  for analyzing
 environmental samples for p-DCB include charcoal  adsorption/desorption.
 extraction with methylene chloride,  purge and trap,  and direct injection
 (EPA 1983a,  1986b,  APHA 1977).  Sample  preparation for biological  samples
 often involves  extraction with hexane  or carbon tetrachloride or  a purge
 and trap technique (Bristol et al.  1982,  EPA  1986c,  Stanley 1986
 Langhorst and Nestrick 1979).

      The GC  separates complex mixtures of organics and allows individual
 compounds to be identified and quantified.

      Detectors  used to identify p-DCB  include the flame ionization
 detector (FID),  electron capture  detector (BCD),  halogen specific
 detector (HSD),  and photoionization  detector  (PID)  (EPA 1983a,  1986b
 Langhorst and Nestrick 1979,  APHA 1977, Jan 1983,  Bristol  et  al.   1982,
 Mottram  et al.  1982).  When unequivocal identification is required,  use
 of  a  mass spectrometer (MS) coupled  to the  GC column (GC-MS)  may be
 employed (EPA 1983a).

 8.1   ENVIRONMENTAL  MEDIA

      Representative methods appropriate for measuring p-DCB in
 environmental media are  listed  in Table 8.1.

 8.1.1  Air

      The American Public Health Association (APHA) method  for
 measurement of organic solvent vapors  in  air  (834) involves adsorption
 of the organic vapors  in the air  onto  an  activated charcoal filter,
 desorption. with carbon disulfide, and  injection of an aliquot of the
 desorbed sample Into a GC equipped with an  FID. The  resulting peaks are
measured and compared with standards.

      Interference may be caused by high temperatures, high humidity,
high sampling flow rates, and other solvents with the same retention
 times. Different GC column materials and  temperatures may be employed to
resolve the interference of other solvents  (APHA 1977).

-------
70    Section  8
     Sod
                         Table 8.1.  Antytfc*!
                                     forf-DCBts

Sample
matnx

Sample
preparation

Analytical
method'
Sample
detection
limit


Accuracy*


References
     Air      Charcoal adsorption,   GC/FID
              desorption with
              carbon disulfide

     Water    Extraction with        GC/MS
              methyiene chloride     (EPA method 623)
              Extraction with        CC/ECD
              methyiene chlonde     (EPA method 612)
                                         25ppbc       90-110%rf    APHA 1977



                                         4 4 «tg/L      63%
                                         1 34 Mg/L    89%
Purge and trap       GC/HISD           0 24 pg/L    97 5%
                    (EPA method 601)
Purge and trap or     GC/HOSD
direct injection       (EPA method 8010)
                          EPA 1983


                          EPA 1983


                          EPA 1983


2.4-300 ppb   42 to 143%    EPA 1986b
     Food     Headspace           GC/FID;
              entrainment          GC/MS
              on Tenax GC
                                        NO*
             ND
                                                                  Mottram et al. 1982
         "GC, gas chromatography; FID. name lonization detector. MS. man spectrometry; ECD. electron cap-
     ture detector. HISD. halide specific detector HOSD, halogen specific detector.
          Average percent recovery.
         'Lowest value for various detected compounds reported during collaborative testing.
          Estimated accuracy of the sampling and analytical method when the personal sampling pump is cali-
     brated with a charcoal tube in the line.
         'Not determined; identified, but not quantified.

-------
                                                 Analytical Methods   71

8.1.2  Vater
     The EPA approved several methods (601, 602, 612,  624,  625)  for
analysis of wastewater samples for p-DCB. All of these methods employ
packed column GC for separation of the organic pollutants,  but differ in
the sample preparation procedures and the detection instrumentation.
     Measuring methods 601 and 612 are optimized for purgeable
halocarbons and chlorinated hydrocarbons, respectively, with detection
limits of about 0.2 to 1.5 Mg/L. Method 625,  which employs  MS for
detection and quantification, is more broadly applicable to a large
number of base/neutral and acid extractable organics.

8.1.3  Soil
     Soil samples can be analyzed for p-DCB by GC with a halogen-
specific detector.  Sample preparation is by purge and trap. This method
is approved by the EPA for analysis of halogenated volatile organics
(8010) in solid waste. The method detection limit for p-DCB ranges from
2.4 to 300 ppb, depending on the matrix (EPA 1986b).

8.1.4  Food
     Analysis of p-DCB in meat is typically done by headspace sampling
and GC/FID or GC/MS analysis (Mottram et al.  1982).

8.2  BIOMEDICAL SAMPLES
     Methods appropriate for measuring p-DCB in biological  samples are
listed in Table 8.2.

8.2.1  Fluids and Exudates
     Blood levels of p-DCB are measured using GC with either an ECD or
PID. The samples are prepared by extraction with hexane or  carbon
tetrachloride (Bristol et al. 1982, Langhorst and Nestrick 1979).
     Analysis of urine for p-DCB is by GC/PID, with sample  extraction
with carbon tetrachloride (Langhorst and Nestrick 1979). As mentioned in
Sect. 2.2.2, the principal method of monitoring for recent p-DCB
exposure is the measurement of 2,5-dichlorophenol in the urine (Pagnatto
and Walkley 1965).
     Human milk may be analyzed  for p-DCB by GC/ECD with extraction by
hexane and concentrated sulfuric acid (Jan 1983) or by GC/MS  (Erickson
et al. 1980).

8.2.2  Tissues
     Adipose tissue levels of p-DCB may be determined by the  same method
used for human milk (Jan 1983) or by a high-resolution gas
chromatography/mass spectroscopy method  (Stanley 1986) which  employs a
dynamic headspace purge and  trap system  for sample preparation.

-------
72     Section  8
                        Table U. Aaalytkil
for p-DCB (•
Sample
matru
Blood


Urine

Human
milk


Adipose



Sample
preparation
Extraction with
hexane
Extraction with
carbon tetrachloride
Extraction with
carbon tetrachloride
Extraction with
hexane/concemrated
sulfunc and

Extraction with
hexane
Dynamic headipace
purge and trap
Analytical
method'
GC/ECD

GC/PD
GC/PD

GLC/ECD


GC/MS
GLC/ECD

HRGC/MS

Sample
detection
limit
2ppb*

30og/g
0.75 ng/g

5Mg/kg*


004ng/mL*
146 Mg/kg'

0.012Mg/g*

Accuracy
82%

89%
81%

>80%


NRC
>80%

94%

Reference*
Bristol el aL 1982

Langhont and
Nestnck 1979
Langhont and
Nestnck 1979
Jan 1983


Enckson et aL 1980
Jan 1983

EPA 1986c

            GC, gai chromatograpby: BCD. electron capture detector, PD. photownizstion detector GLC,
      gas-liouid chramatography; MS, man ipectrometry; HRGC, high-resolution gas chromatography
            Lowest level detected.
            Not reported.
          'Average level detected.

-------
                                                                      73
                   9.  REGULATORY AND ADVISORY STATUS

9.1  INTERNATIONAL

     The World Health Organization  (WHO) has not recommended a drinking
water guideline value for p-DCB based on health effects. The WHO
recommends 0.1 j*g/L p-DCB as a level "unlikely to give rise to taste and
odor problems. ..." (WHO 1984).

9.2  NATIONAL

9.2.1  Regulations

     Regulations applicable to p-DCB address occupational exposure
concentrations, testing requirements, drinking water levels, spill
quantities, and presence in hazardous wastes. Regulations applicable to
p-DCB are summarized in 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
p-DCB is 75 ppm (450 mg/m3) in workplace air for a TWA (8 h/day,  40
h/week).

     The EPA Office of Drinking Water (ODW) has promulgated a Maximum
Contaminant Level (MCL) for p-DCB of 0.075 mg/L. The MCL is normally
based on analyses of health effects and other factors such as the best
available technology, analytical methods, treatment techniques, and
economic factors.

     The Comprehensive Environmental Response,  Compensation and
Liability Act of 1980 (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 RQ
for p-DCB set by the EPA Office of Emergency and Remedial Response
(OERR) is 100 Ib.

     Chemicals are included on the Resource Conservation and Recovery
Act (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. p-DCB is included on this list, and
wastes containing p-DCB are subject to the RCRA regulations promulgated
by the EPA Office of Solid Waste (OSW).

     The Office of Toxic Substances (OTS) promulgates test rules
requiring manufacturers and/or processors of chemicals which may present
an unreasonable risk to health or the environment to conduct tests of
those chemicals for health and/or environmental effects. Manufacturers

-------
74    Seccion  9
                       Tabfe 9.1.
Agency

OSHA

EPA ODW


EPAOERR


EPAOSW

EPAOTS










iPAOPP
Descnption

Permissible exposure limit (PEL) in
workplace air
Tune-weighted average (TWA)
8 h/day, 40 h/week
Maximum contaminant level (MCL)


Reportable quantity (RQ)


Hazardous Constituent List
Appendix VIII

Test rule— requirements for health
effects and chemical fate testing



Preliminary assessment information rate


Health and safety data reporting rule


Comprehensive assessment information
rule (proposed)
Registered pesticide; registration
standard in progress
Vall»e References

75 PP™ 29 CFR 1910
(450mg/mJ) (1971)

0075 mg/L 40 CFR 141 61
52 FR 25690
(07/08/87)
100 Ib 40 CFR 117 3
50 FR 13456
(04/04/85)
NAa 40 CFR 261
45 FR 33084
(05/19/80)
NA 40 CFR 799. 1052
51 FR 11728
(04/07/86)
51 FR 24657
(07/08/86)
NA 40 CFR 712
47 FR 26992
(06/22/82)
NA 40 CFR 716
47 FR 38780
(09/02/82)
NA 5| FR 35762
(10/07/86)
NA 32 FR 588
(01/07/87)
ACGIH        Threshold limit value (TLV)
                TWA
                                                                   ACGIH 1986
                                                     (450 mg/m1)
NIOSH
NIOSH/OSHA
EPA ODW
EPAOWRS
Short-term exposure limit (STEL)
Immediately dangerous to life or health (IDLH)
Respiratory equipment required
Ambient water quality criteria for
prataetwM ft hnn»i| health
Ingesting water and organisms
110 ppm
(665 mg/m1)
1.000 ppm
75 ppm
sad above
0.075 mg/L
0.4 mg/L
2.6 mg/L
NIOSH 1983
NIOSH/OSHA 1981
40 CFR 141.50
52 FR 25690
(07/08/87)
EPA 1980

-------
                                  Regulatory  and Advisory Scacus    75
TtMe9.1 (coMtaed)
Agency

NAS
I ARC
EPA
EPA
Description
EPA ODW
Longer-term HA-child
Longer-term HA-adult
Lifetime HA
Suggested no advene response level (SNARL)
Cancer ranking
Cancer ranking
Carcinogenic potency (q,*)
Value
10.7 mg/L
37 5 mg/L
0075 mg/L
0094 mg/L
Group 2B
Group C
2.2 X I0~2
(mg/kg/day)-'
References

NAS 1977
IARC 1987
52 FR 25690
(07/08/87)
Battelle and
Crump 1986
"NA - not available.

-------
 76   Section 9

 and processors of p-DCB are required to conduct chemical  face and
 reproductive effects tests under current regulations.

      Manufacturers of p-DCB are required to  submit  to  EPA information  on
 the quantity of the 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 p-DCB must be  submitted
 to EPA by manufacturers or processors  under  the Health  and Safety Data
 Reporting Rule.

      The OTS proposed a Comprehensive  Assessment Information  Rule (CAIR)
 which would specify the reporting requirements  for  chemicals 'for  which
 EPA requires information.  This  rule is designed to  streamline  collection
 of information to support  chemical risk assessment/management
 strategies.  p-DCB is one of the 47 chemicals  proposed to  be included in
 this  rule.

      The EPA Office of Pesticide Programs  (OPP)  is  responsible for the
 registration of  all pesticide products sold  in  the  United States. p-DCB
 is  currently a registered  pesticide, and the  OPP expects  to require
 additional  data  when it reviews p-DCB  for  reregistration.  The OPP has
 also  included p-DCB on a list of inert ingredients  of toxicological
 concern  in  pesticide formulations  (52  FR 13305).

 9.2.2  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 p-DCB is  summarized in Table 9.1 and
 includes  the  following:  Immediately Dangerous to Life or  Health (IDLH)
 level  for occupational exposure; a threshold  limit value  (TLV); the
 drinking water Maximum Contaminant Level Goal (MCLG); ambient water
 quality  criteria;  drinking water health  advisories  (HAs);  and a chronic
 Suggested No  Adverse Response Level (SNARL) calculated by  the National
Academy  of  Sciences.

     The NIOSH IDLH for occupational exposure to p-DCB in air is  1000
ppm. This level  represents a  maximum concentration  from which one could
escape within 30  min without  any escape-impairing symptoms or
 irreversible  health effects.  NIOSH/OSHA occupational health guidelines
require  Che use  of minimal respiratory equipment by workers exposed to
p-DCB at 75 ppm  and above.  At levels of  1000  ppm and above, more  complex
and protective equipment is required.

     The American Conference  of Governmental  Industrial Hygienists
 (ACGIH)  recommends  a TLV TWA  of 75  ppm and a  Short-Term Exposure  Limit
 (STEL) of 110  ppm/15 min/8 h  for p-DCB. This  level  is intended to be low
enough to prevent  acute  and chronic poisoning.

     The EPA  ODW MCLG  for  p-DCB, although not an enforceable standard,
 is based on  the health effects  of  p-DCB and is currently  0.075 mg/L.
based on chronic  toxicity  data  and the classification of p-DCB in Group
C (possible human  carcinogen) (52  FR 25690).  The MCLG applies to

-------
                                      Regulatory and  Advisory Scacus    77

 finished  drinking  water.  The  MCL (Maximum Contaminant  Level) has also
 been  set  at  0.075  mg/L.

      The  ODW prepared  HAs for numerous  drinking water  contaminants  The
 HAs describe concentrations of contaminants  in  drinking water at which
 adverse effects would  not be  anticipated  to  occur and  include a margin
 of safety to protect sensitive members  of the population. The HAs are
 calculated for 1-day,  10-day,  longer-term, and  lifetime exposures  For
 p-DCB the longer-term  HA  is 10.7 mg/L for a  child and  37.5 mg/L for an
 adult. The Lifetime HA is 0.075 mg/L. Adequate  data  were not available
 to calculate a 1-day or 10-day HA, but  the EPA  recommends using the
 longer-term  HA for a child for this  duration of exposure (EPA 1987b).

      The  ambient water quality criteria are  guidelines set by the EPA
 Office of Water Regulations and Standards (OWRS) to  protect human health
 from  potential adverse effects from  the ingestion of water and/or
 organisms (fish or invertebrates) or from ingestion  of organisms only
 from  surface water sources. The values  for p-DCB are 0.4 mg/L for
 ingesting water and organisms  and 2.6 mg/L for  ingesting only organisms

 9.2.3  Data  Analysis

      Reference dose.   A reference dose  (RfD) of 0.1  mg/kg/day
 (100  ^gAg/day) has been  calculated  for p-DCB in the final draft of the
Office of Drinking Water  Health Advisory  on  p-DCB (EPA 1987b).  This
value has  not yet been verified by the  Agency's RfD Workgroup.  It is
based on  the NOAEL of  150  mg/kg for  rats  in  the 13-week NTP study,  an
uncertainty  factor of  1000, and a dosing  regimen conversion factor of
 5/7 (5 days  dosing per week).  For further information about the meaning
and calculation of RfDs,  the reader  should consult EPA 1987, Reference
Dose  (RfD): Description and use in health risk  assessments, Appendix A
of the Integrated Risk Information System (IRIS).

      Carcinogenic potency, q.*.  In  an  analysis of the NTP (1987)
carcinogenicity data (available  in 1986 as the  galley draft), Battelle
and Crump  (1986) used  the  liver  tumors  in male  mice and the linearized
multi-stage model to calculate  a q * of 2.2  x 10'2 (mg/kg/day)"*• For a
more detailed explanation  of how q * values  are calculated, the reader
is referred  to Anderson et al.  (1983).

     Therefore, as calculated by EPA  (1987a), lifetime drinking water
exposure  levels corresponding  to 10'6,  10'5, and 10'4 excess risk (based
on 95% upper bound confidence  limit  to  multistage model) would be 1.75,
17.5 and  175 Mg/L,  respectively, for a  70-kg human consuming 2 L of
water per day. There were  not  enough distinct data points to allow fits
to other-models tried  (Weibull,  logit,  probit)  by the EPA's Office of
Drinking Vater.

     Using the male rat kidney  tumor data in the NTP (1987) study with
p-DCB, Battelle and Crump  (1986) reports  a q *  of 6 x 10'3 by the
linearized multistage procedure  as well as by the multistage-Weibull and
Crump's multistage models, taking time  to death into account. This q *
is lower  than the q * of 2.2 x  10'2 obtained with the male mouse liver
tumor data from the NTP (1987)  study and  is, therefore, considered the
less conservative estimate of  risk.  It  should be noted that by using the
linearized multistage models,   the lower limit of risk estimate is zero.

-------
78   Section 9

     A quantitative assessment of risk using the NTP (1987) study has
also been carried out by the Office of Toxic Substances.  The data sets
were modified slightly to eliminate animals that died early during
exposure. The linearized multistage extrapolation model was applied to
the adjusted data. An upper-bound risk of exposure to 1 mg/kg/day was
estimated to equal 2 x 10*2, based on male mouse liver tumors,  and 6 x
10"3, based on male rat kidney tumors.

     The Carcinogen Assessment Group (CAG) considers the model and the
data sets used for the quantitative risk estimates to be appropriate.
The upper-bound value of 2 x 10*2 derived from data on mouse liver
tumors is considered to be a reasonable estimate of risk.  Since chere is
no good evidence to the contrary, when converting to an estimated human
risk from inhalation exposure, it is assumed that equivalent
administered doses will result in equivalent effects. Thus, the risk
from exposure to 1 mg/m3 p-DCB, assuming equivalent absorption
efficiency by either-route, would be estimated to equal 2 x 10~2
mgAg/day x 20 m3/day x 1 mg/m3 divided by 70 kg, or 5.7 x 10'3
mg/m3/day.
     The EPA (1987a) has placed p-DCB in Category C, possible human
carcinogen. This category is for substances with limited evidence of
oncogenic potential in animal studies in the absence of human data.

     Carcinogenic potency, other.  No other estimates of the
carcinogenic potency of p-DCB as developed by other federal agencies
have been located.

9.3  STATE

9.3.1  Regulations
     p-DCB is not specified in any state water quality standards.
However, this compound is included in the category "toxic substances" in
the water quality standards of most states in narrative form. Specific
narrative standards protect use of surface waters for public water
supply, contact recreation, etc.

9.3.2  Advisory Guidance
     Advisory guidance from the states has not been located for p-DCB.

-------
                                                                      79
                             10.  REFERENCES


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

Alexander M, Lustigman BK. 1966. Effects of chemical structure on
microbial degradation of substituted benzenes. J Agric Food Chem
14(4):410-413.

Amoore JE, Hautala E. 1983. Odor as an aid to chemical safety: Odor
thresholds compared with threshold limit values and volatilities for
214 industrial chemicals in air and water dilution. J Appl Toxicol
3:272-290.

Anderson D. 1976. Paradichlorobenzene: Estimation of its mutagenic
potential in the Salmonella, cyphimurium plate incorporation mutagenicity
assay. ICI Report CTL/P/298. November (unpublished, cited in Loeser and
Litchfield 1983).

Anderson D, Hodge MCE. 1976. Paradichlorobenzene: Dominant lethal study
in the mouse. ICI Report CTL/P/296. November (unpublished, cited in
Loeser and Litchfield 1983).

Anderson D, Richardson CR. 1976. Paradichlorobenzene: Cytogenic study in
the rat. ICI Report CTL/P/293. November (unpublished, cited in Loeser
and Litchfield 1983).

Anderson EL et al.  1983. Quantitative approaches in use to assess cancer
risk.  Risk Anal 3(4):377-295.

Anderson KJ, Leighty EG, Takahashi MT. 1972.  Evaluation of herbicides
for possible mutagenic properties. J Agric Food Chem 20:649-656.

APHA (American Public Health Association). 1977. Methods of Air Sampling
and Analysis. 2nd ed. Washington, DC: APHA, pp. 894-902.

Ariyoshi T. Ideguchi K, Iwasaki K, Arakaki M. 1975. Relationship between
chemical structure and activity. II. Influence of isomers of
dichlorobenzene, trichlorobenzene, and tetrachlorobenzene on the
activities of drug-metabolizing enzymes. Chem Pharm Bull 23(4):824-830.
*Key studies.

-------
 80    Section  10

 Astrand  I.  1975. Uptake  of  solvents  in  the blood and tissues of man.
 Scand J  Work  Environ  Health 1:199-208.

 Azouz WM,  Parke DV, Williams RT.  1955.  Studies in detoxication  The
 metabolism of halogenobenzenes. Ortho-  and Paradichlorobenzenes. Biochera
 J 59(3):410-415.

 Ballschmiter  K, Scholz C. 1980. Microbial decomposition of chlorinated
 aromatic substances.  VI.  Formation of dichlorophenols and
 dichloropyrocatechol  from dichlorobenzenes in a micromolar solution by
 Pseudomonas species.  Chemosphere  9(7-8):457-467.

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

 Battelle,  KS Crump and Co.  1986.  Quantitative risk assessment for 1.4-
 dichlorobenzene prepared  for exposure evaluation division. Office of
 Toxic Substances, EPA under Contract 68-02-4246.

 Bombard E, Luckhaus G. Undated. p-Dichlorobenzene:  Subchronic
 toxicological studies on  the subject of nephrotoxic effects (unpublished
 study).

 Bozzelli JW, Kebbekus BB. 1982. A study of some aromatic and halocarbon
vapors in  the ambient atmosphere  of New Jersey. J Environ Sci Health
A17(5):693-711.

 Bramlett JA, Repa EW, Mashni CI.  1986.  Leachate characterization and
 synthetic  leachate formulation for liner testing. In:  HMCRI (Hazardous
Materials Control Research  Institute).  1986.  The  Seventh National
Conference on Management  of Uncontrolled Hazardous  Waste Sites.  December
 1-3,  1986, Washington, DC.

Bristol DV, Crist HL, Lewis RG, MacLeod KE,  Sovocool GW.  1982.  Chemical
analysis of human blood for assessment of environmental exposure to
semivolatile organochlorine chemical contaminants.  J Anal Toxicol
6:269-275.

Brodzinsky R,  Singh HB. Volatile  Organic Chemicals  in Atmosphere:  An
Assessment of Available Data. Contract 68-02-3452.  Environ Sci Res Lab.
Research Triangle Park, NC:  ORD EPA.

Brunce N, Kumar Y,  Ravanal L, Safe S. 1978.  Photochemistry of
chlorinated byphenyls in  iso-octane solution.  J Chem Soc, Perkin
Transact II, pp.  880-884.

Bush  B, Smith RM, Narang AS, Hong C-S.  1984.  Photoionization
conductivity detection limits for environmental pollutants with and
without chromophores. Anal Lett 17:467-474.

-------
                                                          References   81

 Campbell DM, Davidson RJL.  1970. Toxic haemolytic anemia in pregnancy
 due to a pica for paradichlorobenzene. J Obstet Gynaecol Br Commonw
 77:657.

 Carlson GP. 1977. Chlorinated benzene induction of hepatic porphyria
 Experientia 33:1627-1629.

 * Carlson GP.  Tardiff R. 1976. Effect of chlorinated benzenes on the
 metabolism of foreign organic compounds.  Toxicol Appl Pharmacol
 36:383-394.

 Charbonneau M,  Short B,  Lock E,  Swenberg J. 1987.  Mechanism "of
 petroleum-induced sex-specific protein droplet nephropathy and renal
 cell proliferation in Fischer 344 rats:  Relevance  to humans.  Chemical
 Industry Institute of Toxicology, Research Triangle  Park,  NC
 (unpublished study).

 Charbonneau M,  Strassner J,  Lock EA,  Turner MJ,  Swenberg JA.  1987b.
 1,4-Dichlorobenzene-induced nephrotoxicity: Similarity with unleaded
 gasoline  (UG)-induced renal effects.  Third International Symposium  on
 Nephrotoxicity,  Aug.  3-7,  Guilford,  England (in press).

 Chiou  CT, Freed  VH,  Scnmedding DW,  Kohnert RL.  1977.  Partition
 coefficient and  bioaccumulation  of  selected organic  chemicals.  Environ
 Sci Technol 1(5):475-478.

 Clayton GD,  Clayton  FE,  eds.  1981.  Patty's  Industrial Hygiene  and
 Toxicology.  3rd  revised  ed.  Vols. 2A  and  2B. Toxicology. New York: John
 Wiley  and Sons.

 Cone MV, Baldauf MF,  Opresko DM,  Uziel MS.  1983. Chemicals  Identified in
 Human  Breast Milk, a  Literature  Search. Oak Ridge National  Laboratory
 report under EPA/DOE  IAG No.  DW930139-01-1.  Office of Pesticides and
 Toxic  Substances. EPA 560/5-83-009.

 Coniglio WA, Miller K, MacKeever  D. 1980. The occurrence of volatile
 organics in drinking  water.  Briefing  prepared for Deputy Asst. Admin.,
 Drinking Water, March 6. Criteria and Standards Division, EPA,
 Washington, DC.

 Cotter LH.  1953. Paradichlorobenzene  poisoning from  insecticides. NY  J
 Med 53:1690.

 Craig  P. 1986.  Presentations  on male  rat nephropathy. Toxicology Forum
 Aspen,  CO, July 15, 1986.

 Dallas CE, Weir FV, Feldman S, Putcha L,  Bruckner JV. 1983. The uptake
 and distribution of 1,1-dichloroethylene in rats during inhalation
 exposure.  Toxicol Appl Pharmacol 68:140-151.

Elder VA,  Proctor BL, Hites RA. 1981. Organic compounds found near dump
sites  in Niagara Falls,  New York. Environ Sci Technol 15(10):1237-1243.

-------
82   Seccion 10

EPA (Environmental Protection Agency). 1987a. Final Draft Criteria
Document for Ortho-dichlorobenzene, Heta-dichlorobenzene.  Para-
dichlorobenzene.  Criteria and Standards Division, Office of Drinking
Water, EPA, Washington, DC.

EPA (Environmental Protection Agency). 1987b. Health Advisory for
Dichlorobenzenes. Draft. Washington, DC, EPA, Office of Drinking Water

EPA (Environmental Protection Agency). 1986a. Superfund Public Health
Evaluation Manual. Washington, DC, Office of Emergency and Remedial
Response. EPA 540/1-86-060.

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

EPA (Environmental Protection Agency). 1986c. Broad Scale  Analysis of
the FY 82 National Human Adipose Tissue Survey Specimens.  Vol. II.
Volatile Organic Compounds. Office of Toxic Substances, Washington, DC.
EPA 560/5-86-036.

EPA (Environmental Protection Agency). 1985. Health Assessment Document
for Chlorinated Benzenes. Final report. Office of Health and
Environmental Assessment, Washington, DC. EPA/600-8-841015F.

EPA (Environmental Protection Agency). 1983a. Methods for  Chemical
Analysis of Water and Wastes. Office of Research and Development,
Environmental Monitoring and Support Laboratory, Cincinnati, OH.  EPA-
600/4-79-020.

EPA (Environmental Protection Agency). 1983b. Chemicals Identified in
Human Breast Milk. A Literature Search. Office of Pesticides and Toxic
Substances, Washington, DC. EPA 560/5-83-009.

EPA (Environmental Protection Agency). 1981. An Exposure and Risk
Assessment for Dichlorobenzenes. Monitoring and Data Support Division,
Office of Water Regulations and Standards,  EPA,  Washington, DC.

EPA (Environmental Protection Agency). 1980. Ambient Water Quality
Criteria for Dichlorobenzenes. Environmental Criteria and Assessment
Office, Office of Water Regulations and Standards, EPA, Washington, DC
EPA 440/5-80-039.

EPA (Environmental Protection Agency). 1979. Water Related Fate of 129
Priority Pollutants. Vol. II. Monitoring and Data Support Division,
Office of Water Regulations and Standards,  EPA,  Washington, DC.

EPA (Environmental Protection Agency). 1978a. Statement of Basis and
Purpose for an Amendment to the National Interim Primary Drinking Water
Regulations on Trihalomethanes. Office of Water Supply, EPA, Washington.
DC.

-------
                                                          References    83

 EPA (Environmental Protection Agency).  1978b.  In-Depth Studies  on  Health
 and Environmental Impacts of Selected Water Pollutants.  Contract 68-01-
 4646,  EPA,  Washington,  DC.

 EPA (Environmental Protection Agency).  1975.  Identification  of  Organic
 Compounds  in  Effluents  from Industrial  Sources.  Office of Toxic
 Substances, EPA,  Washington,  DC.

 Erickson MD,  Harris  BSH III,  Pellizzari ED,  Tomer KB,  Waddell RD,
 Whitaker DA.  1980. Acquisition and  Chemical  Analysis of  Mother's Milk
 for Selected  Toxic Substances.  EPA  560/13-80-029 (as cited in EPA
 1983b).

 Frank  SB, Cohen HJ.  1961.  Fixed drug  eruption  due to para-
 dichlorobenzene.  NY  Med J  61:4079.

 Gaffney PE. 1976.  Carpet and  rug  industry case study II:  Biological
 effects. J Water  Pollut Control Fed 48(12):2731-2737.

 Gaines TB, Linder RE. 1986. Acute toxicity of  pesticides  in adult and
 weanling rats. Fundam Appl  Toxicol  7:299-308.

 Garrison AW, Hill DW. 1972. Organic pollutants from mill  persist in
 downstream waters. Am Dyestuff  Rep  21-25.

 Ghassemi M, Quinlivan S, Bachmaier J. 1984. Characteristics of  leachates
 from hazardous waste landfills. J Environ Sci Health A19(5):579-620.

 * Giavini E, Broccia ML, Prati  M, Vismara C. 1986.  Teratologic
 evaluation of p-dichlorobenzene in  the  rat. Bull Environ  Contam Toxicol
 37(2):164-168.

 Gupta KC. 1972. Effects  of  some antimitotics on  the cytology of
 Fenugreek roots in vivo  and in  vitro. Cytobios 5(19):179-187.

 Hallowell M. 1959. Acute haemolytic anaemia following  the ingestion of
 paradichlorobenzene. Arch Dis Child 34:74-75.

 Hawkins DR, Chasseaud LF, Woodhouse RN, Cresswell DG.  1980. The
 distribution,  excretion, and biotransformation of p-dichloro-(14)-
 benzene in rats after repeated  inhalation, oral, and subcutaneous doses.
 Xenobiotica. 10:81-95.

 Haworth S,  Lawlor T, Mortelmans K, Speck W, Ziegler E.  1983.  Salmonella
 mutagenicity test results for 250 chemicals. Environ Mutagenesis (Suppl
 1) 5:3-142.

* Hayes WC, Hanley TR Jr, Gushow TS, Johnson KA, John JA. 1985.
Teratogenic potential of inhaled dichlorobenzenes in rats and rabbits.
 Fundam Appl Toxicol 5(1):190-202.

-------
84   Section 10

Herbold B. 1986a. Investigation of p-dichlorobenzene for clastogenic
effects In mice using the micronucleus test. Bayer AG, ed. Institute of
Technology. Report 14694 (unpublished study).

Herbold B. 1986b. Investigation of 2,5-dichlorophenol for clastogenic
effects in mice using the micronucleus test (unpublished study).

* Hodge MCE, Palmer S, Wilson J, Bennett IP. 1977. Paradichlorobenzene
Teratogenicity study in rats. ICI Report CRL/P/340. July 27, 1976
(unpublished, cited in Loeser and Litchfield 1983).

Hollingsworth RL, Rowe VK, Oyen F, Hoyle HR, Spencer HC. 1956.  Toxicity
of paradichlorobenzene: Determinations on experimental animals and human
subjects. AMA Arch Ind Health 14:138-147.

HSDB (Hazardous Substances Data Bank). 1987. Record for 1.4-
Dichlorobenzene. Computer printout. National Library of Medicine  April
9. 1987.

Hull and Co. 1980. Employee Exposure to Chlorobenzene Products.
Greenwich, CT.

IARC (International Agency for Research on Cancer). 1987.  IARC
Monographs on the Evaluation of Carcinogenic Risks to Humans. Overall
Evaluations of Carcinogenicity.  An updating of IARC Monographs Vols. 1
to 42. Suppl 7. IARC (WHO), Lyon, France.

ICF. 1987. Preliminary Use and Substitutes Analysis of Para-
dichlorobenzene. Draft. EPA, Washington, DC.

Institute di Ricerche Biomediche. 1987.  Study of the capacity of the
test article para-dichlorobenzene to induce chromosome aberrations in
human lymphocytes cultured in vitro.  Experiment 1031 (unpublished
study).

Institute di Ricerche Biomediche. 1986b. Study of the capacity of the
test article para-dichlorobenzene to induce gene mutation in V79 Chinese
hamster lung cells. Experiment 1030 (unpublished study).

Institute di Ricerche Biomediche. 1986a. Study of the capacity of the
test article para-dichlorobenzene to induce "unscheduled DNA synthesis"
in cultured HeLa cells. Experiments M1032/1-2 (unpublished study).

Irie D et al. 1973. Acute toxicity, inhalation toxicity, and skin
irritation of cyclododecane, tricyclododecane, naphthalene, and p-
dichlorobenzene (parazol.) Toho Igakkai Zasshi 20:772 (Japanese;
abstract in English).

Jan J. 1983. Chlorobenzene residues in human fat and milk. Bull Environ
Contain Toxicol 30:595-599.

Jerina DM, Daly JW. 1974. Science 1985:573-582.

-------
                                                          References    85

 Kanerva RL,  Ridder GM,  Lefever FR,  Alden CL.  1987.  Comparison of shore-
 term renal effects due  to oral administration of decalin or  d-limonene
 in young adult male Fisher-344 rats.  Food Chem Toxicol  25(5):345-353.

 Kimura R.  Hayashi  T,  Sato M,  Aimoto T.  Murata T.  1979.  Identification of
 sulfur-containing  metabolites of p-dichlorobenzene  and  their  disposition
 in rats.  J Pharm Dyn 2:237-244.

 Kitamura R,  Sumino SK,  Mio T.  1977.  Metabolic pathway of chlorobenzenes
 Koenshu-Iyo  Masu Kenkyukai 2:79-88.

 Kopperman HL,  Keuhl DW,  Glass GE. 1976.  Chlorinated compounds  found in
 waste  treatment effluents and their capacity  to bioaccumulate.
 Proceedings  of the Conference on the  Environmental  Impact of Water
 Chlorination,  Oak  Ridge,  TN,  October  22-24.

 Langhorst  ML.  Nestrick  TJ.  1979.  Determination of chlorobenzenes  in air
 and biological samples  by gas  chromatography  with photoionization
 detection. Anal Chem  51:2018-2025.

 Langner  HJ,  Hillinger HG.  1971.  Taste variation of  the  egg caused by the
 deodorant  p-dichlorobenzene.  Analytical  proof.  Berlin.  Muenchen
 Tierairztl 84:851  (German).

 Lebret E,  Van  de Wiel H,  Bos  H,  Noij D,  Boleij, J.  1986.  Volatile
 organic  compounds  in  Dutch homes. Environ Int 12:323-332.

 Leo A, Hansch  C, Elkins  D. 1971.  Partition coefficients and their uses
 Chem Rev 71:525-616.

 Litton Bionetics.  1986. Mutagenicity evaluation of  2.5-dichloro-phenol
 in  the CHO HGPRT forward  mutation assay  (unpublished study).

 Litton Bionetics.  1985. Evaluation of 2,5-dichlorophenol  in the in vitro
 transformation  of  BALB/3T3 cells assay  (unpublished study).

 * Loeser E, Litchfield MH. 1983. Review  of recent toxicology studies on
 p-dichlorobenzene.   Food Chem Toxicol 21:825-832.

 Lowenheim  FA, Morgan  MK.  1975. Faith, Keyes,  and Clark's  Industrial
 Chemicals. 4th  ed.

 Lu P. Metealf RL.  1975. Environmental fate and  biodegradability of
 benzene derivatives as studied in a model  aquatic ecosystem. Environ
 Health Perspect 10:269-284.

 Miranda CL, Want JL,  Henderson MC, Nakaue HS, Buhler DR.  1984. Effects
 of chlorobenzenes on hepatic porphyrin and drug metabolism in chick
 embryo and day-old chick. Res Commun Chem  Pathol Pharmacol 46(l):13-24.

Monsanto.  1986. Material  safety data sheet for  Santochlor (para-
 dichlorobenzene). Monsanto Company,  St.  Louis, MO.

-------
 86    Section  10

 Morita M,  Mimura S.  Ohi G.  Yagyu H.  Nlshizawa T.  1975. A  systematic
 determination of chlorinated benzenes  in human adipose tissue   Environ
 Pollut 9:175-179.

 Morita M.  Ohi G.  1975.  Paradichlorobenzene  in human  tissue and
 atmosphere in Tokyo  metropolitan area. Environ Pollut 8:267-274.

 Mottram DS, Edwards  RA,  MacFie  HJH.  1982. A comparison of the flavour
 volatiles  from cooked beef  and  pork  meat systems. J  Sci Food Aerie
 33:934-944.                                                   B

 NAS  (National Academy of Sciences).  1977. Drinking Water and Health.
 National Academy  of  Sciences, Washington, DC.

 Neptune D.  1980.  Descriptive  statistic for detected priority pollutants
 and  tabulation listings.  Off  Water Plan Stand, EPA, Washington,  DC
 TRDB-0280-001.

 NFPA  (National Fire  Protection  Association). 1978. Fire Protection Guide
 for Hazardous  Materials.

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

 NIOSH/OSHA. 1981. Occupational  Health Guidelines for Chemical Hazards,
 p-Dichlorobenzene. DHHS  (NIOSH)  Publication 81-123, January 1981.

 NIOSH  1980. Health Hazard Evaluation Report. PPG Industries,  Natrium  WV
 80-082-773.

 * NTP  (National Toxicology Program).  1987.  Toxicology and Carcinogenesis
 Studies of 1,4-Dichlorobenzene  (CAS No. 106-46-7) in F344/N Rats and
 B6C3F1 Mice (Gavage Studies). NTP TR 319.  NIH Publication 87-2575.

Oliver BG, Nicol KD. 1982. Chlorobenzenes in sediments,  water, and
 selected fish  from Lakes Superior, Huron,  Erie,  and Ontario.  Environ Sci
Technol 16(8):532-536.

OSHA (Occupational Safety and Health Administration). 1986.  Computer
printout OSHA Computerized Information System (OCIS), SIC IH Information
 File for p-Dichlorobenzene. Salt Lake City,  Utah: OSHA,  U.S.  Department
of Labor.

Pagnatto LD, Walkley JE. 1965. Urinary dichlorophenol as  an index of
paradichlorobenzene exposure. Am Ind Hyg Assoc J 26:137-142.

Perocco P, Bolognesl S,  Alberghini W. 1983.  Toxic activity of seventeen
 industrial solvents and halogenated compounds on human lymphocytes
cultured in vitro. Toxicol Lett  16(1-2):69-75.

-------
                                                         References   87

Perrin H. 1941. Possible hannfulness of paradichlorobenzene used as a
moth killer. Bull de 1'Acad de Med 125:302 (French; translated to
English).

Petit G, Champeix J. 1948. Does an intoxication caused by para-
dichlorobenzene exist? Arch des Malad Prof de Med 9:311 (French;
translated to English).

Prasad I, Pramer D. 1968. Mutagenic activity of some chloroanilines and
chlorobenzenes. Genetics 20:212-213.

Prasad I. 1970. Mutagenic effects of the herbicide 3,4-dichloro-
proprionanilide and its degradation products. Can J Microbiol
16:369-372.

Preston BD,  Miller JA, Miller EC. 1983. Non-arene oxide aromatic ring
hydrocyclation of 2,2',5.5'-tetrachlorobiphenyl as the major metabolic
pathway catalyzed by phenobarbital-induced rat liver microsomes. J Biol
Chem 258(13):8304-8311.

Rautio AW. 1988. Chlorobenzene Producers Association comments on the
Draft Toxicological Profile for 1,4-Dichlorobenzene. Submitted to the
Agency for Toxic Substances and Disease Registry, March 7, 1988.

* Riley RA,  Chart IS, Doss A. Gore CU, Patton D, Weight TM. 1980.
Para-dichlorobenzene: Long-term inhalation study in the rat. ICI Report
CTL/P/447. August 1980 (unpublished, cited in Loeser and Litchfield
1983).

Rimington GE,  Ziegler G. 1963. Experimental porphyria in rats induced by
chlorinated benzenes. Biochem Pharmacol 12:1387-1397.

Sax NI. 1979.  Dangerous Properties of Industrial Materials. Sth ed. NY:
Van Nostrand Reinhold.

Schmidt GE.  1971. Abnormal odor and taste due to p-dichlorobenzene. Arch
Lebensmittelhyg 22:43  (German abstract).

Scuderi R. 1986. Determination of Para-dichlorobenzene Releases from
Selected Consumer Products. Midwest Research Institute Project 8801-A
for EPA Contract 68-02-4252. Draft final report. October 23, 1986.

Sharma AK, Sarkar SK.  1957. A study of the comparative effect of
chemicals on chromosomes of roots, pollen mother cells, and pollen
grains. Proc Ind Acad  Sci B XLV(XXX):288-293.

Sharma AK, Battacharya NK. 1956. Chromosome breakage through para-
dichlorobenzene treatment. Cytologia 21:353-360.

Simmon VF, Riccio ES,  Pierce MV. 1979. In vitro microbiological
genotoxicity tests of  chlorobenzene, rn-dichlorobenzene, o-dichlorobenzene,
and p-dichlorobenzene. Unpublished report by SRI International for EPA.
Contract 68-02-2947. Final report. May 1979.

-------
 88   Section 10

 Singh HB,  Salas LJ.  Smith AJ.  Shlgeishl H.  1981.  Measurements of some
 potentially hazardous organic  chemicals In  urban  atmospheres   Atmos
 Environ 15(4) .-601-612.

 Srivastava LM.  1966.  Induction of mitotic abnormalities  in certain
 genera of  tribe Vicieae by paradichlorobenzene. Cytologia  31:166.

 Steinmetz  KL, Spanggord RJ.  1987a.  Examination of the  potential  of
 p-dichlorobenzene to  induce  unscheduled DNA synthesis  or DNA  replication
 in the in  vivo-in vitro mouse  hepatocyte DNA  repair assay  (unpublished
 study).

 Steinmetz  KL, Spanggord RJ.  1987b.  Evaluation of  the potential of
 p-dichlorobenzene to  induce  unscheduled DNA synthesis  or DNA  replication
 in the in  vivo-in vitro rat  kidney  DNA  repair assay (unpublished study)

 Swenberg,  J.  1987. Personal  communication.  June 9, 1987.

 Symons JM,  Bellar TA, Caldwell JK,  et al. 1975. National organics
 monitoring survey for halogenated organics.  J Am Waterworks Assoc
 67:634-636.

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

 Wallace L,  Pellizzari E,  Sheldon  L, Hartwell  T, Sparacino C,  and Zelon
 H.  1986. The  total exposure  assessment methodology (TEAM) study: Direct
 measurement of  personal  exposures through air and water  for 600
 residents  of  several U.S.  cities. In: Cohen Y, ed. Pollutants in a
 Multimedia Environment.   Plenum Publishing.

 Wallgren K. 1953. Chronic  intoxications  in  the manufacture of moth
 proofing agents consisting of  mainly paradichlorobenzene. Zentralbl
 Arbeitsmed Arbeitsschutz  (Darmstadt) 3:14-15.

 Ware SA, Weast  WL. 1977.  Investigation of selected potential
 environmental contaminants:  Halogenated benzenes.  Environmental
 Protection Agency, Office  of Toxic  Substances, Washington,  DC. EPA
 560/2-77-004.

 Weast  RC,  ed. 1985. CRC Handbook of Chemistry and Physics.  66th  ed.
 Boca Raton, FL: CRC Press.

 Weller RW,  Crellin AJ. 1953. Pulmonary granulomatosis following
 extensive  use of paradichlorobenzene. Arch Intern Med 91:408.

 Wetzel R,  Durst C, Sarno D,  et  al.  1985. Demonstration of in situ
 biological degradation of  contaminated groundwater and soils.  In: HMCRI
 (Hazardous Materials Control Research Institute).  1985. The Sixth
National Conference on Management of Uncontrolled Hazardous Waste Sites.
November 4-6, 1985, Washington, DC.

-------
                                                         References   89

WHO (World Health Organization). 1984. Guidelines for Drinking Water
Quality. Vol. 1. Recommendations. Geneva, Switzerland: World Health
Organization.

Williams RT. 1959. The metabolism of halogenated aromatic hydrocarbons.
In: Detoxication Mechanisms. 2nd ed. John Wiley and Sons, New York
pp. 237-258.

-------
                                                                       91
                              11.  GLOSSARY

 Acute Exposure-•Exposure to a chemical for a duration of 14 days or
 less, as specified in the 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 Efface Level  (FEL)--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.

-------
 92   Section  11

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

 Immune-logic Toxicity--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)  (LD50)--The  dose of a chemical which has been calculated
 to cause death in  50% of a defined experimental animal population.

 Lovest-Observed-Adverae-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   93

Neurotoxicity- -The occurrence of adverse effects on Che 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 to
calculate an estimate of carcinogenic potency, the incremental excess
cancer risk per unit of exposure (usually /ig/L for water, mg/kg/day for
food, and pg/n^ 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. HI of the Clean Water
Act. Quantities are measured over a 24-h period.

Reproductive Toxicity--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.

-------
 94    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  the
 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.

-------
                                                                      95
                         APPENDIX:  PEER REVIEW

     A peer review panel was assembled for p-DCB. The panel consisted of
the following members: Dr. C. Klaassen, University of Kansas Medical
Center; Dr. S. Safe, Texas A&M University; and Mr. L. Skory (retired),
Dow Chemical Company.  These experts collectively have knowledge of
p-DCB's physical and chemical properties, toxicokinetics, key health end
points, 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 Act 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 list 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 to
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.

-------