Download Cancer Statistics

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Prostate-specific antigen wikipedia , lookup

Transcript
Cancer Statistics
in Metropolitan Detroit 2012
Metropolitan Detroit Cancer Surveillance System
Surveillance, Epidemiology and End Results Program
December 2012
Table of Contents
Table of Contents
1
Metropolitan Detroit Cancer Surveillance System (MDCSS)
2
Geographic Coverage Area 3
Population Estimates 4
Michigan Department of Community Health Collaboration,
and MDCSS Authority to Collect Cancer Data
5
Cancer Trends, Incidence, Mortality and Survival in Metropolitan Detroit
6
Temporal Cancer Trends 7
Cancer Incidence and Mortality: All Races
8
Cancer Incidence and Mortality: Whites
10
Cancer Incidence and Mortality: African-Americans
12
Survival Analysis by Stage: Prostate Cancer 14
Survival Analysis by Stage: Female Breast Cancer 15
Survival Analysis by Stage: Lung Cancer 16
Survival Analysis by Stage: Colorectal Cancer 17
Cancer Research at MDCSS
18
The Young Women’s Health History Study Breast Cancer Risk Factors in Young Women
19
Risk Factors for Ovarian Cancer in African-American Women 20
Patterns of Care Study 21
Genetic Pathways of Inflammation and Risk of Lung Cancer
and Chronic Obstructive Pulmonary Disease
22
The Epidemiology Research Core
24
List of Studies Conducted at the MDCSS
25
MDCSS/SEER Publications 2010-2012
28
Affiliations and Acknowledgements
32
MDCSS Participating Institutions
33
Appendix A: Michigan Public Health Code and Health
Insurance Portability and Accountability Act (HIPAA) Excerpts
34
Credits36
Metropolitan Detroit Cancer Surveillance System (MDCSS)
and Surveillance, Epidemiology and End Results (SEER) Cancer
Program Description and Goals
2
Program Description
Program Goals
The Metropolitan Detroit Cancer Surveillance System (MDCSS)
has been recording data on cancer in Wayne, Oakland and Macomb
Counties since 1973. Cancer is a reportable disease under State
of Michigan law. The MDCSS is the arm of the Michigan Department of Community Health (MDCH) responsible for the collection
and reporting of cancer data for residents of the tri-county area.
The MDCSS is also one of 20 cancer registries in the United States
providing cancer data to the National Cancer Institute as part of the
Surveillance, Epidemiology and End Results (SEER) Program.
The SEER Program was established pursuant to the National Cancer
Act of 1971. The MDCSS is a founding participant in that program
and is now completing its 40th year of data collection as a SEER
Registry.
The national SEER Program collects cancer data on approximately
28% of the United States population. Participating registries are
chosen in part for the special populations represented within their
borders. The Detroit tri-county area is an ideal location because of
its urban, industrial setting and diverse population. The goals of the
Detroit SEER Program are to:
The MDCSS has a staff of more than 50 people who collect data
from hospitals, laboratories, radiation centers and other facilities
that serve cancer patients. A follow-up program tracks more than
95% of the cancer survivors diagnosed since 1973, providing yearly
updates on their vital status. Funding for the registry is provided by
the National Cancer Institute to Wayne State University. Additional
funding for research studies is provided by the National Cancer
Institute and other agencies through grants and contracts and
distributed to university, state and national researchers investigating cancer-related topics. Strict rules are in place to preserve the
confidentiality of individual patient information and the hospitals
and physicians who provide these data.
1. Assemble and report estimates of cancer incidence, survival and
mortality in the tri-county area.
2. Monitor annual cancer trends to identify unusual changes in
specific forms of cancer in population subgroups.
3. Provide information on changes over time in the extent of disease
at diagnosis, therapy and patient survival.
4. Promote and conduct studies designed to identify factors leading
to cancer causes, prevention and control.
5. Respond to requests from individuals and organizations for data
on cancer in the tri-county area.
Over the past 40 years, we have seen remarkable improvements in
cancer diagnosis and therapy. Early detection leads to improved survival. In fact, mortality from cancer has decreased in recent years,
which is most likely due to early detection and better therapies.
Geographic Coverage Area:
Metropolitan Detroit
The MDCSS and the Metropolitan Detroit SEER Program
provide reporting for Wayne,
Oakland and Macomb counties in southeastern Michigan.
These three counties are home
to 3,863,924 people, approximately 39% of the total state
population. There are 1,820,584
individuals residing in Wayne
County, 39% of those in the City
of Detroit, which has a population of 713,777. Oakland County residents number 1,202,362
and Macomb County has the
smallest population of the
three, with 840,978 residents.
About 69% of the population in
the tri-county area are White
and 26% African-American. An
additional 5% of the population
are of other races, primarily
Asian and Native American or
are multi-racial. (U.S. Census
2010.) As of the 2010 census,
persons of Hispanic ethnicity
made up about 4% of the total
population. The tri-county area
also has great ethnic diversity,
which includes groups of Polish-, German-, Italian-, and
Arab-Americans.
In 2010, there were 24,651 new
invasive and in situ cancers
diagnosed in residents of the
metropolitan Detroit (tricounty) area. This is about half
the number reported for the
entire state. Because of the
large population encompassed
in the metropolitan Detroit
area and the diversity found
here, the MDCSS is especially
vital to both State of Michigan
and national cancer statistics
reporting.
Newly Diagnosed Invasive and In Situ Cancers
in the Metropolitan Detroit Area (2010)
Cancers
11,314 New
Wayne County
Cancers
7,797 New
Oakland County
Cancers
5,540 New
Macomb County
3
Population by Age, Sex, Race and Ethnicity
in Metropolitan Detroit, 2011
OAKLAND
MACOMB
WAYNE
4
AMERICAN INDIAN/
WHITEAFRICAN-AMERICAN
ALASKA NATIVE
AGE
ASIAN/PACIFIC
HISPANIC*
ISLANDER
ALL RACES
MALE
FEMALEMALEFEMALE MALEFEMALEMALEFEMALEMALEFEMALEMALEFEMALE
00-04 years 74,443 71,227 37,033 35,689
783
760 6,291 5,934 9,101 8,736118,550113,610
05-09 years 82,624 78,660 36,392 34,865
777
819 6,915 6,755 8,921 8,739126,708121,099
10-14 years 89,203 84,559 40,276 39,271
847
844 6,264 5,988 7,966 7,714136,590130,662
15-19 years 87,808 81,699 44,995 43,715
889
923 5,211 4,599 7,346 6,890138,903130,936
20-24 years 77,771 75,432 38,494 41,051
782
757 4,373 4,410 6,263 5,875121,420121,650
25-29 years 79,629 78,017 27,689 32,977
673
671 5,369 5,894 6,455 6,027113,360117,559
30-34 years 80,248 78,917 26,928 34,046
652
764 6,109 7,331 6,770 6,365113,937121,058
35-39 years 80,373 79,405 29,062 37,611
696
739 7,062 7,992 6,221 5,975117,193125,747
40-44 years 95,525 94,753 30,739 38,345
750
836 6,972 6,879 5,426 5,118133,986140,813
45-49 years 104,613104,665 29,734 36,356
742
808 5,821 5,636 4,496 4,434140,910147,465
50-54 years 110,011112,777 30,812 38,277
706
824 4,562 4,499 3,708 3,652146,091156,377
55-59 years 98,606100,855 27,536 35,233
623
687 3,611 3,895 2,776 3,028130,376140,670
60-64 years 82,404 87,034 22,902 30,490
428
556 2,986 3,726 2,104 2,146108,720121,806
65-69 years 56,38562,51114,652 19,365 238 325 2,341 2,499 1,321 1,403 73,616 84,700
70-74 years 38,43947,291 9,451 13,642 195 226 1,665 1,629 817 98949,75062,788
75-79 years
29,004
40,223
6,809
11,413104145892
1007634888
36,809
52,788
80-84 years
25,007
38,145
4,973
9,024 53123451666521718
30,484
47,958
85+ years 20,716
44,071
4,072
9,425 53144310506389607
25,151
54,146
Total
1,312,809
1,360,241
462,549540,795 9,99110,95177,20579,84581,23579,304
1,862,554
1,991,832
Source: Surveillance, Epidemiology, and End
Results (SEER) Program (www.seer.cancer.gov)
SEER*Stat Database: Populations -
*Note: Persons of Hispanic origin may be of any race, therefore Hispanic counts not added to totals.
Total U.S. (1990-2011) <Katrina/Rita Adjustment>
- Linked To County Attributes - Total U.S., 19692011 Counties, National Cancer Institute, DCCPS,
Surveillance Research Program, Surveillance
Systems Branch, released October 2012.
Michigan Department of Community Health Collaboration
and the MDCSS Authority to Collect Cancer Data
The 28-year collaboration
between the State of Michigan
Department of Community
Health (MDCH) and the Metropolitan Detroit Cancer Surveillance System (MDCSS) was
recently renewed by the State of
Michigan. The following letter
grants authority to the MDCSS
to collect cancer data as a designated public health authority of
the State. Because the MDCSS
is designated by the MDCH as
a “state-authorized entity for
the collection and reporting of
individually identifiable cancer
information,” HIPAA allows
hospitals to disclose such information to the MDCSS.
5
Cancer Trends, Incidence, Mortality
and Survival in Metropolitan Detroit
6
Temporal Cancer Trends 7
Cancer Incidence and Mortality: All Races
8
Cancer Incidence and Mortality: Whites
10
Cancer Incidence and Mortality: African-Americans
12
Survival Analysis by Stage: Prostate Cancer 14
Survival Analysis by Stage: Female Breast Cancer 15
Survival Analysis by Stage: Lung Cancer 16
Survival Analysis by Stage: Colorectal Cancer 17
Temporal Cancer Trends in Metropolitan Detroit
Lung cancer has been the
leading cause of cancer
mortality among males since
1973, but it has declined
somewhat since 1992.
Mortality among females
from lung cancer continues to
increase, reflecting an increase
in smoking. Mortality rates
for prostate and female breast
cancer have been decreasing
over time.
Age-Adjusted Incidence for Invasive Lung & Bronchus,
Prostate & Female Breast Cancers, Metropolitan Detroit, 1973 – 2009
RATES PER 100,000 POPULATION
300
300
250
250
200
200
150
150
100
100
50
50
1975
1980
1985
1990
1995
2000
2005
Source: Surveillance, Epidemiology, and End Results (SEER) Program (www.seer.cancer.gov) SEER*Stat Database: Incidence - SEER 9 Regs
Research Data, Nov 2011 Sub, Vintage 2009 Pops (1973-2009) <Katrina/Rita Population Adjustment> - Linked To County Attributes - Total
U.S., 1969-2010 Counties, National Cancer Institute, DCCPS, Surveillance Research Program, Surveillance Systems Branch, released April
2012, based on the November 2011 submission.
Female Lung
Male Lung
Prostate
Female Breast
Age-Adjusted Mortality Rates for Invasive Lung & Bronchus,
Prostate & Female Breast Cancers, Metropolitan Detroit, 1973 – 2009
RATES PER 100,000 POPULATION
Incidence rates for prostate
cancer peaked in 1992, but
have remained high due to
increased detection through the
PSA (prostate specific antigen)
test. Female breast cancer
incidence rates have remained
steady since the late 1980’s.
Mammography screening has
played an important role in the
early diagnosis of this cancer.
While rates of lung cancer have
been declining among males
since the mid 1990’s, rates
among females are only now
leveling off.
100
100
80
80
60
60
40
40
20
20
1975
1980
1985
1990
1995
2000
2005
Source: Surveillance, Epidemiology, and End Results (SEER) Program (www.seer.cancer.gov) SEER*Stat Database: Mortality - All COD,
Aggregated With State, Total U.S. (1969-2009) <Katrina/Rita Population Adjustment>, National Cancer Institute, DCCPS, Surveillance
Research Program, Surveillance Systems Branch, released April 2012. Underlying mortality data provided by NCHS (www.cdc.gov/nchs).
7
All Races
Cancer Incidence, Mortality and Age Adjusted Rates per 100,000
Population by Site, Race and Sex, Metropolitan Detroit, 2009
8
All Races
Incidence
Rate
Mortality
Rate
Total MaleFemale
Total MaleFemale
Total MaleFemale
Total MaleFemale
All Sites (Invasive)
21,97211,22810,744508.3593.3451.47,9494,0133,936184 224.3158.8
Oral Cavity and Pharynx
518
345
173
11.9
17.2
7.3
90
60
30
2.1
3.1
1.2
Lip
14
7
7
0.3
0.4
0.3
*
*
0
*
*
0
Tongue
152
102
50
3.4
5
2.1
17
11
*
0.4
0.6
*
Salivary Gland
59
33
26
1.4
1.8
1.1
*
*
*
*
*
*
Floor of Mouth
38
25
13
0.9
1.2
0.6
*
*
0
*
*
0
Gum and Other Mouth
82
44
38
1.9
2.3
1.6
14
*
*
0.3
*
*
Nasopharynx
19
13
6
0.4
0.6
0.2
11
*
*
0.3
*
*
Tonsil
71
60
11
1.6
2.8
0.5
*
*
*
*
*
*
Oropharynx
27
19
8
0.6
0.9
0.3
10
*
*
0.2
*
*
Hypopharynx
37
30
7
0.9
1.6
0.3
*
*
*
*
*
*
Other Oral Cavity
and Pharynx
19
12
7
0
0.5
0
16
*
*
0.4
*
*
Digestive System
3,9972,0981,89991.7 111.177.2 2,0401,111929 46.8 60.8 36.7
Esophagus 24716879 5.6 8.8 3.1 18413153 4.2 7 2.1
Stomach 3512211308.1 12.25.4 16695 71 3.9 5.5 2.8
Small Intestine
109
50
59
3
2.8
2.4
12
*
*
0.3
*
*
Colon and Rectum
1,993
950
1,043
46
50.6
42.7
759
374
385
17.5
21.2
14.9
Colon excluding Rectum1,432
65278033.235.331.864130533614.917.513.1
Cecum
333
131
202
7.8
7.2
8.3
†
†
†
†
†
†
Appendix
30
15
15
0.7
0.8
0.7
†
†
†
†
†
†
Ascending Colon
287
121
166
6.7
6.6
6.7
†
†
†
†
†
†
Hepatic Flexure
81
36
45
1.9
2
1.8
†
†
†
†
†
†
Transverse Colon
113
52
61
2.6
2.7
2.4
†
†
†
†
†
†
Splenic Flexure
40
22
18
0.9
1.1
0.7
†
†
†
†
†
†
Descending Colon
80
46
34
1.9
2.6
1.5
†
†
†
†
†
†
Sigmoid Colon
370
185
185
8.5
9.7
7.6
†
†
†
†
†
†
Large Intestine, NOS
98
44
54
2.3
2.5
2.2
†
†
†
†
†
†
Rectum and
56129826312.815.410.911869 49 2.7 3.7 1.8
Rectosigmoid Junction
Rectosigmoid Junction
134
72
62
3.1
3.7
2.6
†
†
†
†
†
†
Rectum
427
226
201
9.7
11.6
8.3
†
†
†
†
†
†
Anus, Anal Canal
80
34
46
1.8
1.8
1.8
*
*
*
*
*
*
and Anorectum
Liver and Intrahepatic
Bile Duct 34626086 7.5 12.73.4 25418074 5.7 9 3
Liver 32424678 7.1 11.93.1 20814959 4.6 7.4 2.4
Intrahepatic Bile Duct
22
14
8
0.5
0.8
0.3
46
31
15
1
1.6
0.6
Gallbladder 6221411.41.21.625 * 220.6* 0.8
Other Biliary
84
45
39
1.9
2.4
1.6
19
10
*
0.4
0.6
*
Pancreas 66633033615.317.613.658930028913.516.311.6
Retroperitoneum
10
7
*
0.2
0.4
*
*
*
*
*
*
*
Peritoneum, Omentum
and Mesentery
26
*
25
0.6
*
1
*
*
*
*
*
*
Other Digestive Organs
23
11
12
1
0.6
1
12
*
*
0.3
*
*
Respiratory System
3,5751,8651,71084 101.971.5 2,3371,2701,06754.8 70.3 44
Nose, Nasal Cavity
and Middle Ear
28
19
9
0.7
1
0.4
*
*
*
*
*
*
Larynx 184146384 7.62 4737101.12 0.4
Lung and Bronchus
3,3261,6741,65278.3 91.7 69.1 2,2761,2231,05353.3 67.7 43.4
Pleura
31
23
8
0.7
1.3
0.3
*
*
0
*
*
0
Trachea, Mediastinum and
Other Respiratory Organs
6
*
*
0.1
*
*
*
*
*
*
*
*
Bones and Joints
32
18
14
0.8
0.9
0.7
19
12
*
0.4
0.6
*
Soft Tissue including Heart
12861673.13.23 5533221.31.81
Skin excluding Basal
and Squamous 75141533617.522.114.811777 40 2.6 4.2 1.6
Melanoma of the Skin
639
351
288
14.9
18.7
12.7
91
61
30
2.1
3.3
1.2
Other Non-Epithelial Skin
11264483 3.52 2616100.60.90.4
Breast
3,123
21
3,102
72
1.2
131.3
614
*
606
14.1
*
24.6
All Races
Incidence
Rate
Mortality
Rate
Total MaleFemale
Total MaleFemale
Total MaleFemale
Total MaleFemale
Female Genital System 1,309
01,309
29.6
054.7
383
0383
8.8
015.7
Cervix Uteri 174
0174
4.2
07.9
490491.2
02.2
Corpus and Uterus, NOS
707
0
707
15.7
0
28.9
128
0
128
2.9
0
5.2
Corpus Uteri
685
0
685
15.2
0
28
59
0
59
1.4
0
2.5
Uterus, NOS
22
0
22
0.5
0
0.9
69
0
69
1.6
0
2.7
Ovary 3210 3217.3 0 13.31820 1824.2 0 7.4
Vagina
26
0
26
0.6
0
1.1
*
0
*
*
0
*
Vulva 640 641.50 2.6140 140.30 0.5
Other Female Genital Organs 17
0
17
0
0
0.8
*
0
*
*
0
*
Male Genital System
3,689
3,689
0
84
188.9
0
358
358
0
8.2
21.7
0
Prostate
3,5633,5630
81 182.10
353 353 0
8.1 21.5 0
Testis
102
102
0
2.7
5.5
0
*
*
0
*
*
0
Penis
19
19
0
0.4
1
0
*
*
0
*
*
0
Other Male Genital Organs
*
*
0
*
*
0
0
0
0
0
0
0
Urinary System
1,8351,266569 42.368.823.2410 268 142 9.6 15.35.7
Urinary Bladder 1,006
75025623.242.110.221514471 5 8.4 2.8
Kidney and Renal Pelvis
778
486
292
17.9
25
12.2
186
119
67
4.4
6.7
2.8
Ureter
30
16
14
0.7
1
0.5
*
*
*
*
*
*
Other Urinary Organs
21
14
7
0.5
0.8
0.3
*
*
*
*
*
*
Eye and Orbit
23
12
11
0.6
0.7
0.4
*
*
*
*
*
*
Brain and Other
Nervous System 2661371296.4 7.2 5.8 18310182 4.2 5.2 3.4
Brain
241
124
117
5.8
6.6
5.3
†
†
†
†
†
†
Cranial Nerves Other
Nervous System
25
13
12
0.6
0.7
0.5
†
†
†
†
†
†
Endocrine System 61915046914.87.7 21.438 16 22 0.9 0.8 0.9
Thyroid
593
137
456
14.2
7
20.8
23
*
16
0.5
*
0.6
Other Endocrine
including Thymus
26
13
13
0.6
0.7
0.6
15
*
*
0.4
*
*
Lymphoma 99252646623.628.220.13081691397.2 9.7 5.6
Hodgkin’s Lymphoma
130
65
65
3.3
3.5
3.1
17
*
11
0.4
*
0.5
Hodgkin’s - Nodal
124
62
62
3.1
3.4
2.9
†
†
†
†
†
†
Hodgkin’s - Extranodal
6
*
*
0.2
*
*
†
†
†
†
†
†
Non-Hodgkin’s
Lymphoma (NHL) 86246140120.324.717 2911631286.8 9.4 5.1
NHL - Nodal
597
331
266
14
17.8
11.2
†
†
†
†
†
†
NHL - Extranodal
265
130
135
6.3
6.9
5.9
†
†
†
†
†
†
Myeloma 3151821337.39.85.416483 81 3.84.53.2
Leukemia 50429520912 16.29 3301771537.8 10.36
Lymphocytic Leukemia 22114576 5.37.93.383 48 35 1.92.81.4
Acute Lymphocytic
Leukemia
44
27
17
1.2
1.4
0.9
11
*
*
0.3
*
*
Chronic Lymphocytic
Leukemia 16010456 3.85.82.368 38 30 1.62.31.2
Other Lymphocytic
Leukemia
17
14
*
0.4
0.7
*
*
*
*
*
*
*
Myeloid and Monocytic
Leukemia 2571361216.17.55.216184 77 3.84.83.2
Acute Myeloid Leukemia
155
77
78
3.7
4.3
3.4
132
74
58
3.2
4.3
2.4
Acute Monocytic Leukemia
*
0
*
*
0
*
*
0
*
*
0
*
Chronic Myeloid Leukemia
93
54
39
2.2
2.9
1.6
19
*
14
0.4
*
0.5
Other Myeloid/Monocytic
Leukemia
8
*
*
0.2
*
*
*
*
*
*
*
*
Other Leukemia 2614120.60.80.58645412 2.71.5
Other Acute Leukemia
10
*
7
0.2
*
0.3
30
16
14
0.7
1
0.5
Aleukemic, Subleukemic
and NOS
16
11
*
0.4
0.6
*
56
29
27
1.3
1.7
1
Miscellaneous 2961481486.7 8.1 5.7 49826922911.415.38.9
Rates are per 100,000 and age-adjusted to the 2000 US Std Population (19 age groups - Census P25-1130) standard.
† Not reported
* Statistic not displayed due to fewer than 6 cases.
Source: Surveillance, Epidemiology, and End Results (SEER) Program (www.seer.cancer.gov) SEER*Stat Database. Underlying mortality data provided by NCHS (www.cdc.gov/nchs).
9
Whites
Cancer Incidence, Mortality and Age Adjusted Rates per 100,000
Population by Site, Race and Sex, Metropolitan Detroit, 2009
10
WHITE
IncidenceRateMortality Rate
Total MaleFemale
Total MaleFemale
Total MaleFemale
Total MaleFemale
All Sites (Invasive)
16,4048,3678,037501.4573.8455.55,9153,0082,907177 215.2153.6
Oral Cavity and Pharynx
383
254
129
11.7
16.6
7.3
62
40
22
1.9
2.8
1.2
Lip
13
7
6
0.4
0.5
0.3
*
*
0
*
*
0
Tongue
118
82
36
3.6
5.3
2
15
10
*
0.5
0.7
*
Salivary Gland
46
25
21
1.5
1.9
1.2
*
*
0
*
*
0
Floor of Mouth
27
19
8
0.8
1.1
0.5
*
*
0
*
*
0
Gum and Other Mouth
63
35
28
1.9
2.5
1.6
12
*
*
0.4
*
*
Nasopharynx
12
8
*
0.4
0.5
*
*
*
*
*
*
*
Tonsil
53
44
9
1.6
2.7
0.5
*
*
0
*
*
0
Oropharynx
16
10
6
0.5
0.7
0.3
*
*
*
*
*
*
Hypopharynx
23
18
*
0.7
1.3
*
*
*
*
*
*
*
Other Oral Cavity
and Pharynx
12
6
6
0.3
0.3
0.3
11
*
*
0.3
*
*
Digestive System
2,7921,4971,29583.5 102.569 1,448812 636 42.9 56.9 32.6
Esophagus 19214250 5.79.52.515211240 4.57.62
Stomach 226153736.810.7
3.99867313 4.81.6
Small Intestine
66
35
31
2
2.4
1.6
*
*
*
*
*
*
Colon and Rectum 1,419
69572442.848 38.954026827215.919.213.5
Colon excluding Rectum1,013
47553830.433.228.545421424013.415.712
Cecum
234
98
136
7.1
6.9
7.1
†
†
†
†
†
†
Appendix
21
11
10
0.7
0.8
0.6
†
†
†
†
†
†
Ascending Colon
209
88
121
6.3
6.1
6.3
†
†
†
†
†
†
Hepatic Flexure
59
27
32
1.8
2
1.6
†
†
†
†
†
†
Transverse Colon
83
35
48
2.4
2.3
2.5
†
†
†
†
†
†
Splenic Flexure
23
15
8
0.6
1
0.4
†
†
†
†
†
†
Descending Colon
49
29
20
1.5
2.1
1.1
†
†
†
†
†
†
Sigmoid Colon
267
141
126
8.1
9.7
6.8
†
†
†
†
†
†
Large Intestine, NOS
68
31
37
2
2.2
2
†
†
†
†
†
†
Rectum and
Rectosigmoid Junction
406
220
186
12.3
14.8
10.4
86
54
32
2.5
3.6
1.5
Rectosigmoid Junction
86
43
43
2.6
2.9
2.4
†
†
†
†
†
†
Rectum
320
177
143
9.7
11.9
8.1
†
†
†
†
†
†
Anus, Anal Canal
and Anorectum
62
23
39
1.8
1.5
2.1
*
*
*
*
*
*
Liver and Intrahepatic
Bile Duct 20715849 6 10.22.5 15911346 4.7 7.5 2.5
Liver 18514441 5.49.22.212389 34 3.75.91.9
Intrahepatic Bile Duct
22
14
8
0.6
1
0.4
36
24
12
1.1
1.6
0.7
Gallbladder 4114271.21 1.419 * 160.5* 0.7
Other Biliary
63
33
30
1.8
2.3
1.6
15
*
*
0.4
*
*
Pancreas 46622624013.915.512.643422920512.916 10.8
Retroperitoneum
10
7
*
0.3
0.5
*
*
*
*
*
*
*
Peritoneum, Omentum
and Mesentery
22
*
21
0.7
*
1.1
*
*
*
*
*
*
Other Digestive Organs
18
10
8
0.6
0.8
0.4
*
*
*
*
*
*
Respiratory System
2,6831,3731,31082.4 97 72.4 1,759928 831 53.4 66.1 44.8
Nose, Nasal Cavity
and Middle Ear
21
15
6
0.6
1.1
0.3
*
*
*
*
*
*
Larynx
130
100
30
4
6.9
1.7
26
23
*
0.8
1.6
*
Lung and Bronchus
2,4981,2341,26476.787.369.91,722897 825 52.363.944.6
Pleura
31
23
8
0.9
1.6
0.4
*
*
0
*
*
0
Trachea, Mediastinum and
Other Respiratory Organs
*
*
*
*
*
*
*
*
0
*
*
0
Bones and Joints
27
15
12
0.9
1.1
0.8
15
*
*
0.5
*
*
Soft Tissue including Heart
10245573.43.23.54429151.42 1
Skin excluding Basal
and Squamous 71439831622.327.319.411375 38 3.3 5.2 2.1
Melanoma of the Skin
624
349
275
19.5
23.8
17.1
88
59
29
2.6
4.1
1.6
Other Non-Epithelial Skin
90
49
41
2.7
3.4
2.3
25
16
*
0.7
1.1
*
Breast
2,319
16
2,303
70.8
1.2
132.3
430
*
425
12.8
*
22.6
WHITE
IncidenceRateMortality Rate
Total MaleFemale
Total MaleFemale
Total MaleFemale
Total MaleFemale
Female Genital System 9970 99730 0 56.72860 2868.5 0 15.4
Cervix Uteri 113
0113
3.8
07.4
270270.9
01.7
Corpus and Uterus, NOS
552
0
552
16.2
0
30.6
89
0
89
2.6
0
4.8
Corpus Uteri
545
0
545
15.9
0
30.2
43
0
43
1.3
0
2.4
Uterus, NOS 7070.2
00.4
460461.4
02.4
Ovary 2500 2507.5 0 14.11490 1494.4 0 7.9
Vagina
16
0
16
0.5
0
1
*
0
*
*
0
*
Vulva 530 531.60 2.9120 120.40 0.6
Other Female Genital Organs 13
0
13
0.4
0
0.8
*
0
*
*
0
*
Male Genital System
2,589
2,589
0
78.6
172.5
0
248
248
0
7.2
18.8
0
Prostate 2,478
2,478
074.5
164.1
0243
243
0718.5
0
Testis
90
90
0
3.5
6.8
0
*
*
0
*
*
0
Penis
16
16
0
0.5
1.1
0
*
*
0
*
*
0
Other Male Genital Organs
*
*
0
*
*
0
0
0
0
0
0
0
Urinary System
1,4731,037436 44.372 23.7343 224 119 10.316.26.4
Urinary Bladder 86765721025.846.611 17912356 5.3 9 2.9
Kidney and Renal Pelvis
560
351
209
17.1
23.3
11.9
156
97
59
4.7
7
3.3
Ureter
28
16
12
0.8
1.2
0.6
*
*
*
*
*
*
Other Urinary Organs
18
13
*
0.6
0.9
*
*
*
*
*
*
*
Eye and Orbit
18
8
10
0.5
0.6
0.5
*
*
*
*
*
*
Brain and Other
11
Nervous System 20511095 6.77.85.815285 67 4.65.83.7
Brain
186
100
86
6.1
7.1
5.2
†
†
†
†
†
†
Cranial Nerves Other
Nervous System
19
10
9
0.6
0.7
0.5
†
†
†
†
†
†
Endocrine System 48112435715.98.4 23.328 10 18 0.8 0.7 1
Thyroid
467
118
349
15.5
8
22.8
19
*
13
0.5
*
0.7
Other Endocrine
including Thymus
14
6
8
0.4
0.4
0.4
*
*
*
*
*
*
Lymphoma 80843037825.730.422.12611441177.8 10.56
Hodgkin’s Lymphoma
104
53
51
3.8
4.1
3.5
14
*
*
0.4
*
*
Hodgkin’s - Nodal
98
50
48
3.5
3.8
3.3
†
†
†
†
†
†
Hodgkin’s - Extranodal
6
*
*
0.3
*
*
†
†
†
†
†
†
Non-Hodgkin’s
Lymphoma (NHL) 70437732721.926.318.62471391087.3 10.15.5
NHL - Nodal
486
265
221
15.1
18.5
12.4
†
†
†
†
†
†
NHL - Extranodal
218
112
106
6.8
7.8
6.1
†
†
†
†
†
†
Myeloma 19111378 5.88.14.197 48 49 2.83.42.3
Leukemia 40124116012.617.19.3 2551421137.8 10.75.7
Lymphocytic Leukemia 18011961 5.88.53.763 38 25 1.93 1.2
Acute Lymphocytic
Leukemia
35
20
15
1.4
1.5
1.2
*
*
*
*
*
*
Chronic Lymphocytic
Leukemia 13186454 6.12.45332211.62.51.1
Other Lymphocytic
Leukemia
14
13
*
0.4
0.9
*
*
*
*
*
*
*
Myeloid and Monocytic
Leukemia 20111190 6.27.85.111868 50 3.65 2.7
Acute Myeloid Leukemia
123
65
58
3.8
4.6
3.3
98
61
37
3.1
4.5
2
Acute Monocytic Leukemia
*
0
*
*
0
*
0
0
0
0
0
0
Chronic Myeloid Leukemia
71
41
30
2.2
2.8
1.6
12
*
*
0.3
*
*
Other Myeloid/Monocytic
Leukemia
6
*
*
0.2
*
*
*
*
*
*
*
*
Other Leukemia 20 11 9 0.60.80.574 36 38 2.22.81.8
Other Acute Leukemia
9
*
6
0.3
*
0.3
27
14
13
0.8
1.1
0.6
Aleukemic, Subleukemic
and NOS
11
8
*
0.3
0.6
*
47
22
25
1.4
1.7
1.1
Miscellaneous 2211171046.4 8.2 5.2 36920816110.915 8.2
Rates are per 100,000 and age-adjusted to the 2000 US Std Population (19 age groups - Census P25-1130) standard.
† Not reported
* Statistic not displayed due to fewer than 6 cases.
Source: Surveillance, Epidemiology, and End Results (SEER) Program (www.seer.cancer.gov) SEER*Stat Database. Underlying mortality data provided by NCHS (www.cdc.gov/nchs).
African-Americans
Cancer Incidence, Mortality and Age Adjusted Rates per 100,000
Population by Site, Race and Sex, Metropolitan Detroit, 2009
12
AFRICAN-AMERICANIncidence
Rate
Mortality
Rate
Total MaleFemale
Total MaleFemale
Total MaleFemale
Total MaleFemale
All Sites (Invasive)
5,0262,5952,431544.1687.3448.41,951971 980 221.7280.2185.3
Oral Cavity and Pharynx
125
85
40
12.8
19.9
7.5
28
20
*
3
4.7
*
Lip *0 **0 *000000
Tongue
32
19
13
3.2
4.4
2.3
*
*
*
*
*
*
Salivary Gland
11
6
*
1.2
1.3
*
*
*
*
*
*
*
Floor of Mouth
9
*
*
1.1
*
*
0
0
0
0
0
0
Gum and Other Mouth
18
9
9
1.8
1.8
1.7
*
0
*
*
0
*
Nasopharynx
*
*
*
*
*
*
*
*
0
*
*
0
Tonsil
18
16
*
1.8
3.8
*
*
*
*
*
*
*
Oropharynx
11
9
*
1.1
2.1
*
*
*
*
*
*
*
Hypopharynx
14
12
*
1.5
2.9
*
*
*
0
*
*
0
Other Oral Cavity
and Pharynx
6
*
*
0.6
*
*
*
*
*
*
*
*
Digestive System 1,106
552554122149.1
103.9
56528627963.981.252.2
Esophagus 5426286 7.65.13118133.55.22.3
Stomach 113644913.2
209.46226367.38.56.8
Small Intestine
42
15
27
4.9
5
5.1
*
*
*
*
*
*
Colon and Rectum 51922729257.461.154.921410411025.331.821.1
Colon excluding Rectum
380
156
224
42.9
43.7
42.5
182
89
93
21.5
27.1
17.9
Cecum
91
27
64
10.6
8.1
12.5
†
†
†
†
†
†
Appendix
7
*
*
0.8
*
*
†
†
†
†
†
†
Ascending Colon
75
31
44
8.6
8.6
8.5
†
†
†
†
†
†
Hepatic Flexure
21
9
12
2.3
2.4
2.2
†
†
†
†
†
†
Transverse Colon
27
15
12
3
4.3
2.2
†
†
†
†
†
†
Splenic Flexure
16
6
10
1.9
1.7
1.9
†
†
†
†
†
†
Descending Colon
28
16
12
3.1
4.7
2.2
†
†
†
†
†
†
Sigmoid Colon
88
37
51
9.4
9.6
9.4
†
†
†
†
†
†
Large Intestine, NOS
27
13
14
3.1
3.8
2.6
†
†
†
†
†
†
Rectum and
Rectosigmoid Junction139716814.6
17.4
12.4
3215173.94.73.1
Rectosigmoid Junction
44
27
17
4.9
7.1
3.3
†
†
†
†
†
†
Rectum
95
44
51
9.7
10.3
9.1
†
†
†
†
†
†
Anus, Anal Canal
and Anorectum
17
10
7
1.7
2.3
1.2
*
*
*
*
*
*
Liver and Intrahepatic
Bile Duct 123903312.1
20.7
5.78762259 14.8
4.6
Liver 123903312.1
20.7
5.78057238.213.6
4.2
Intrahepatic Bile Duct
0
0
0
0
0
0
*
*
*
*
*
*
Gallbladder
20
6
14
2.2
1.5
2.6
*
0
*
*
0
*
Other Biliary
18
11
7
2.1
3.1
1.5
*
*
*
*
*
*
Pancreas
193 102 91 21.527.717.2149 69 80 16.518.814.8
Retroperitoneum 000000 **0 **0
Peritoneum, Omentum
and Mesentery
*
0
*
*
0
*
0
0
0
0
0
0
Other Digestive Organs
*
*
*
*
*
*
*
*
*
*
*
*
Respiratory System 84346537893.6126.9
71.156333422964 94 44.4
Nose, Nasal Cavity
and Middle Ear
*
*
*
*
*
*
*
*
0
*
*
0
Larynx
51
43
8
5.1
10.9
1.3
21
14
*
2.3
3.8
*
Lung and Bronchus 78441636887.6114.5
69.553931822161.389.742.7
Pleura 000000 **0 **0
Trachea, Mediastinum and
Other Respiratory Organs
*
*
*
*
*
*
*
0
*
*
0
*
Bones and Joints
*
*
*
*
*
*
*
*
*
*
*
*
Soft Tissue including Heart
24
15
9
2.4
3.5
1.6
11
*
*
1.2
*
*
Skin excluding Basal
and Squamous
31
15
16
3.3
3.9
2.9
*
*
*
*
*
*
Melanoma of the Skin
10
*
9
1
*
1.6
*
*
*
*
*
*
Other Non-Epithelial Skin
21
14
7
2.2
3.7
1.3
*
0
*
*
0
*
Breast
715
*
710
76.5
*
129.4
175
*
172
19.3
*
31.8
AFRICAN-AMERICANIncidence
Rate
Mortality
Rate
Total MaleFemale
Total MaleFemale
Total MaleFemale
Total MaleFemale
Female Genital System 280
0280
29.5
050.5
9309310.6
017.8
Cervix Uteri 550 555.70 9.9200 202.20 3.7
Corpus and Uterus, NOS
142
0
142
15
0
25.5
39
0
39
4.6
0
7.5
Corpus Uteri
127
0
127
13.4
0
22.9
16
0
16
1.9
0
3.1
Uterus, NOS
15
0
15
1.5
0
2.6
23
0
23
2.7
0
4.4
Ovary 600 606.40 10.9
310 313.70 6.1
Vagina
10
0
10
1.1
0
1.8
*
0
*
*
0
*
Vulva
10
0
10
1.1
0
1.8
*
0
*
*
0
*
Other Female Genital Organs
*
0
*
*
0
*
0
0
0
0
0
0
Male Genital System 9899890 104.9
2560 1081080 13.337.20
Prostate 9769760 103.5
2530 1081080 13.337.20
Testis 1010012.3
0000000
Penis **0 **0000000
Other Male Genital Organs
000000000000
Urinary System
321 201 120 35.556.921.864 42 22 7.5 12.54.1
Urinary Bladder 12280421424.2
7.73520154.25.92.8
Kidney and Renal Pelvis
195
120
75
21.1
32.3
13.7
28
21
*
3.2
6.2
*
Ureter *0 **0 *000000
Other Urinary Organs
*
*
*
*
*
*
*
*
0
*
*
0
Eye and Orbit
*
*
*
*
*
*
0
0
0
0
0
0
Brain and Other
13
Nervous System 4821274.94.95 2915143 3.32.7
Brain
43
18
25
4.4
4.1
4.7
†
†
†
†
†
†
Cranial Nerves Other
Nervous System
*
*
*
*
*
*
†
†
†
†
†
†
Endocrine System
103
15
88
10.5
4
15.6
*
*
*
*
*
*
Thyroid
93
10
83
9.4
2.6
14.6
*
*
*
*
*
*
Other Endocrine
including Thymus
10
*
*
1.1
*
*
*
*
*
*
*
*
Lymphoma 153837016.2
20.9
12.9
4525204.96.73.8
Hodgkin’s Lymphoma
18
8
10
1.8
1.9
1.8
*
*
*
*
*
*
Hodgkin’s - Nodal
18
8
10
1.8
1.9
1.8
†
†
†
†
†
†
Hodgkin’s - Extranodal
0
0
0
0
0
0
†
†
†
†
†
†
Non-Hodgkin’s
Lymphoma (NHL) 135756014.3
1911.1
4224184.66.53.4
NHL - Nodal
97
58
39
10.2
14.7
7.1
†
†
†
†
†
†
NHL - Extranodal
38
17
21
4.1
4.3
4
†
†
†
†
†
†
Myeloma 121685313.3
17.8
10.1
6535307.49.55.9
Leukemia 8745429.912.7
8 6932377.89 7
Lymphocytic Leukemia 3420143.85.32.72010102 2.31.8
Acute Lymphocytic
Leukemia
9
7
*
0.9
1.4
*
*
*
0
*
*
0
Chronic Lymphocytic
Leukemia
23
12
11
2.7
3.6
2.1
15
*
*
1.5
*
*
Other Lymphocytic
Leukemia
*
*
*
*
*
*
*
0
*
*
0
*
Myeloid and Monocytic
Leukemia 4722255.46.64.83713244.54 4.7
Acute Myeloid Leukemia
2811173.33.43.32810183.43.23.5
Acute Monocytic Leukemia
0
0
0
0
0
0
*
0
*
*
0
*
Chronic Myeloid Leukemia
17
11
6
1.9
3.2
1.1
*
*
*
*
*
*
Other Myeloid/Monocytic
Leukemia
*
0
*
*
0
*
*
*
0
*
*
0
Other Leukemia
6
*
*
0.7
*
*
12
*
*
1.3
*
*
Other Acute Leukemia
*
0
*
*
0
*
*
*
*
*
*
*
Aleukemic, Subleukemic
and NOS
*
*
*
*
*
*
*
*
*
*
*
*
Miscellaneous 7130418 8.27.7121586314.2
1811.8
Rates are per 100,000 and age-adjusted to the 2000 US Std Population (19 age groups - Census P25-1130) standard.
† Not reported
* Statistic not displayed due to fewer than 6 cases.
Source: Surveillance, Epidemiology, and End Results (SEER) Program (www.seer.cancer.gov) SEER*Stat Database. Underlying mortality data provided by NCHS (www.cdc.gov/nchs).
Survival Analysis of Prostate Cancer by Stage
Over the past 31 years, the
treatment of men with prostate
cancer diagnosed at an early
stage has improved. In the
tri-county area, the current
five-year relative survival for
men diagnosed with local stage
prostate cancer is near 100%.
Dramatic improvement in
survival of men with prostate
cancer that is diagnosed after
it has spread to other parts of
the body has not been seen.
The five-year relative survival
for men diagnosed with distant
stage disease has fluctuated
for the last 31 years between
20-38%, with African-American
men showing a better trend in
the last decade than White men.
Armed with successful treatment for early stage disease,
there has been a concerted
effort to identify these cancers
while they are in local stage.
Currently, about 95% of the
men who are newly diagnosed
with prostate cancer in the
tri-county area are diagnosed at
local or regional stages. A few
of the risk factors for developing the disease are age, race
(African-American), and family
Localized/Regional
14
100
Distant
Invasive Prostate Cancer – White Males
Stage Distribution 1973 – 2009 Metropolitan Detroit
100
6080
6080
60
40
60
40
40
20
40
20
20
20
1975
1975
1980
1980
1975
1985
1990
1995
2000
2005
1975
1980
1980
1985
1990
1995
2000
2005
YEAR DIAGNOSED - 3 YEAR MOVING AVERAGE
YEAR DIAGNOSED - 3 YEAR MOVING AVERAGE
5-Year Relative Survival Rates for Prostate Cancer –
White Males by Stage, 1973 – 2009 Metropolitan Detroit
5 -Year Relative Survival Rates for Prostate Cancer –
African-American Males by Stage, 1973 – 2009 Metropolitan Detroit
100
80
80
PERCENTAGE
100
PERCENTAGE
Invasive Prostate Cancer – African-American Males
Stage Distribution 1973 – 2009 Metropolitan Detroit
PERCENTAGE
PERCENTAGE
100
80
PERCENTAGE
PERCENTAGE
100
80
100
history (men with close blood
relatives such as fathers or
brothers who have been diagnosed with the disease).
60
40
20
60
40
20
1975
1975
1980
1980
2000
1985
1990
1995
2000
YEAR DIAGNOSED - 3 YEAR MOVING AVERAGE
2005
1975
1980
1985
1990
1995
2000
YEAR DIAGNOSED - 3 YEAR MOVING AVERAGE
2005
Survival Analysis of Female Breast Cancer by Stage
Both diagnosis and treatment
for female breast cancer have
improved since 1973. Due to
mammography screening, more
cases are diagnosed at an earlier
stage. Currently, five-year relative survival for cases diagnosed
at local stage is 95% for women
in the general population. For
women with regional stage
disease, survival also has improved. For women diagnosed
with late stage disease, survival
has remained relatively steady,
at 10-20% for women in the
general population. AfricanAmerican women tend to have a
higher proportion of late stage
diagnoses and five-year survival,
regardless of stage, tends to be
poorer than for White women.
Gender and age are the greatest risk factors for developing
breast cancer. Breast cancer can
Regional
Localized
100
strike men and women both,
but women are many times
more likely to develop the disease than men. A family history
of breast cancer has also been
linked to increasing a woman’s
likelihood of developing the
disease. Gender, age, and family
history are things that cannot
be prevented, however women
can increase their chances of
surviving this disease by con-
Distant
Invasive Breast Cancer – White Females
Stage Distribution 1973 – 2009 Metropolitan Detroit
80
80
PERCENTAGE
100
PERCENTAGE
15
Invasive Breast Cancer – African-American Females
Stage Distribution 1973 – 2009 Metropolitan Detroit
100
60
40
20
60
40
20
1975
1980
1985
1990
1995
2000
2005
1975
YEAR DIAGNOSED - 3 YEAR MOVING AVERAGE
1980
1985
1990
1995
2000
2005
YEAR DIAGNOSED - 3 YEAR MOVING AVERAGE
5-Year Relative Survival Rates for Breast Cancer –
5-Year Relative Survival Rates for Breast Cancer –
African-American Females by Stage, 1973 – 2009 Metropolitan Detroit
100 White Females by Stage, 1973 – 2009 Metropolitan Detroit
100
80
80
PERCENTAGE
100
PERCENTAGE
sulting with their physician, and
following recommended screening for their specific age and risk
group. Women who are at an
increased risk due to a family
history should consult with
their doctor about whether they
should begin mammography
screening at an earlier age.
60
40
20
60
40
20
1975
1980
1985
1990
1995
2000
YEAR DIAGNOSED - 3 YEAR MOVING AVERAGE
2005
1975
1980
1985
1990
1995
2000
YEAR DIAGNOSED - 3 YEAR MOVING AVERAGE
2005
Survival Analysis of Lung Cancer by Stage
Diagnosis of lung cancer in
local stage is difficult because
symptoms usually do not appear
until the disease is advanced.
Only about 23% of lung cancers in Whites and 18-19% in
African-Americans are diagnosed at the local stage. Most
early stage lung cancer cases
are diagnosed incidentally. The
five-year survival rate for local
stage cancer is less than 50% for
the general population. The five-
year survival for distant stage
cancer is approximately 2%. An
effective method for screening
for lung cancer has not yet been
developed although promising
methods are being tested. When
a new method is developed,
the hope is that it will be as
effective and universally used
as mammography screening
has been for breast cancer and
the PSA blood test has been for
prostate cancer.
Regional
Localized
16
100
The major risk factor for lung
cancer is tobacco smoking.
The risk increases over time
as a person continues to smoke.
However, if a person is able
to quit smoking before cancer
has developed, the lung tissue
can begin to repair itself. An
ex-smoker can lower his risk of
lung cancer, but his risk will still
be higher than that of someone
who has never smoked. Nonsmokers who are exposed to
Distant
Invasive Lung Cancer – White Population
Stage Distribution 1973 – 2009 Metropolitan Detroit
Invasive Lung Cancer – African-American Population
Stage Distribution 1973 – 2009 Metropolitan Detroit
80
80
60
60
PERCENTAGE
100
PERCENTAGE
100
40
40
20
20
1975
1980
1985
1990
1995
2000
2005
1975
YEAR DIAGNOSED - 3 YEAR MOVING AVERAGE
1980
1985
1990
1995
2000
2005
YEAR DIAGNOSED - 3 YEAR MOVING AVERAGE
5-Year Relative Survival Rates for Lung Cancer –
100 White Population by Stage, 1973 – 2009 Metropolitan Detroit
5-Year Relative Survival Rates for Lung Cancer –
African-American Population by Stage, 1973 – 2009 Metropolitan Detroit
100
80
80
60
60
PERCENTAGE
100
PERCENTAGE
second hand smoke in the home
or workplace are also at increased risk for developing lung
cancer. Exposure to carcinogens
in the workplace or the environment and having a family
history of lung cancer have
also been found to contribute
to an increased risk for getting
the disease.
40
40
20
20
1975
1980
1985
1990
1995
2000
YEAR DIAGNOSED - 3 YEAR MOVING AVERAGE
2005
1975
1980
1985
1990
1995
2000
YEAR DIAGNOSED - 3 YEAR MOVING AVERAGE
2005
Survival Analysis of Colorectal Cancer by Stage
The five-year survival rate for
colorectal cancer has gradually
increased over the past 31 years
for the general population.
Currently, the five-year relative
survival rate for local stage
cancer is over 90% compared to
73% in 1973. At distant stage,
the five-year relative survival
rate is over 10% compared to
4% in 1973. This improved
survival has been linked to improved early detection practices
including the increased use of
screening sigmoidoscopy.
Age, family history, and diet
are risk factors in developing
colorectal cancer. The symptoms
of colorectal cancer may mimic
some other conditions such as
hemorrhoids or inflammatory
Regional
Localized
100
bowel disease. It is best to consult with a physician to determine the correct cause of the
symptoms. Screening typically
begins at age 50 for the general
population. If there is family
history of the disease, a patient
needs to confer with their physician to discuss when screening
should begin.
Distant
Invasive Colorectal Cancer – White Population
Stage Distribution 1973 – 2009 Metropolitan Detroit
Invasive Colorectal Cancer – African-American Population
Stage Distribution 1973 – 2009 Metropolitan Detroit
6080
80
60
40
60
PERCENTAGE
100
PERCENTAGE
PERCENTAGE
80
100
40
20
20
40
20
1985
1975
1980
1985
1990
1995
2000
2005
1980
1985
1990
1990
1995
1995
2000
2000
2005
YEAR DIAGNOSED - 3 YEAR MOVING AVERAGE
YEAR DIAGNOSED - 3 YEAR MOVING AVERAGE
5-Year Relative Survival Rates for Colorectal Cancer –
5-Year Relative Survival Rates for Colorectal Cancer –
African-American Population by Stage, 1973 – 2009 Metropolitan Detroit
100 White Population by Stage, 1973 – 2009 Metropolitan Detroit
80
100
PERCENTAGE
PERCENTAGE
1975
100
80
60
40
60
PERCENTAGE
6080
40
20
20
40
20
1975
1980
1985
1990
1995
2000
YEAR DIAGNOSED - 3 YEAR MOVING AVERAGE
2005
1975
1980
1985
1990
1995
2000
YEAR DIAGNOSED - 3 YEAR MOVING AVERAGE
2005
17
Cancer Research at MDCSS
18
The Young Women’s Health History Study - Breast Cancer Risk Factors in Young Women
19
Risk Factors for Ovarian Cancer in African-American Women 20
Patterns of Care Study 21
Genetic Pathways of Inflammation and Risk
of Lung Cancer and Chronic Obstructive
Pulmonary Disease
22
The Young Women’s Health History Study Breast Cancer Risk Factors in Young Women
Ellen Velie, PhD (Principal Investigator), Kendra Schwartz, MD, MSPH
Ellen Velie, PhD at Michigan State University, with Co-Investigators,
Kendra Schwartz, MD, MSPH at Wayne State University (WSU)
and Ann Hamilton, PhD at the University of Southern California
(USC), assisted by Westat, a research corporation, are conducting
the “Young Women’s Health History Study”, funded by the National
Cancer Institute, to evaluate risk factors for breast cancer in young
women. The study is being conducted in a socioeconomically diverse
population of African-American and White women in Metropolitan
Detroit and Los Angeles County. The study’s objectives are to
examine whether potentially modifiable lifestyle factors measured
over a woman’s lifetime, related genetic factors, as well as the social
conditions that influence lifestyle factors, explain why some young
women develop specific types of breast cancer. Researchers plan to
enroll 4,000 young women – both those who have had breast cancer
and those who have had not, to compare these two groups of women.
The “Young Women’s Health History Study” began recruitment of
participants in December of 2011. Young women randomly selected
for this study are encouraged to participate. Whether or not women
have had breast cancer, their story will give researchers a fuller
picture of women’s lives. Unfortunately, due to the studies’ scientific
methodology, women who are not contacted cannot volunteer
themselves. For questions regarding this research, please visit the
study web-site, http://ywhhs.org/. In the metropolitan Detroit area
you can also contact the study toll-free phone number: 1-(877)679-9687 and either Dr. Kendra Schwartz or the Study Project
Coordinator, Ms. Kathy Cross will get back with you.
Age-Adjusted Incidence of Invasive Female Breast Cancer
in Women (50+), SEER-9, 1992 – 2009
RATE PER 100,000
400
400
300
300
1992
1995
1998
2001
2004
2007
YEAR OF DIAGNOSIS
Age-Adjusted Incidence of Invasive Female Breast Cancer
in Women (<40), SEER-9, 1992 – 2009
20
RATE PER 100,000
In the United States, White women are more likely to develop breast
cancer after age 50 (figure 1), but among the youngest women,
under age 40, African American women are more likely to develop
breast cancer (figure 2)3. African American and poorer women also
appear more likely to develop tumors with the worst prognosis3,4.
In addition, compared to similarly aged White women, AfricanAmerican women have higher death rates from breast cancer (figure
3). In young African-American, under 40 years of age, only about 42%
of breast cancers are diagnosed at the local stage, before the cancer
has spread beyond the breast, whereas in young White women 48%
are diagnosed before the cancer has spread5. Screening is not usually
recommended before age 40 because current screening modalities do
not work well for young women. It’s therefore particularly important
we understand the causes of breast cancer in young women, in order
to prevent the development of the disease.
500
16
12
8
19
1992
1995
1998
2001
2004
2007
YEAR OF DIAGNOSIS
Surveillance, Epidemiology, and End Results (SEER) Program (www.seer.cancer.gov) SEER*Stat
Database: Incidence - SEER 13 Regs Research Data, Nov 2011 Sub, Vintage 2009 Pops (1992-2009)
<Katrina/Rita Population Adjustment> - Linked To County Attributes - Total U.S., 1969-2010 Selection restricted to SEER 9 registries only.
White
African-American
Age-Adjusted Mortality of Invasive Female Breast Cancer
in Young Women (<40), SEER-9, 1973 – 2009
66
RATE PER 100,000
Each year in the United States, about 44,000 women are newly
diagnosed with breast cancer before 50 years of age (SEER)1. Tumors
in these young women are more likely to be aggressive with a worse
prognosis, but their causes remain poorly understood2.
55
44
33
22
11
0
1975
1980
1985
1990
1995
2000
2005
YEAR OF DEATH
Reference:
1.Howlader N, Noone AM, Krapcho M, Neyman N, Aminou R, Altekruse SF, Kosary
CL, Ruhl J, Tatalovich Z, Cho H, Mariotto A, Eisner MP, Lewis DR, Chen HS, Feuer
EJ, Cronin KA (eds). SEER Cancer Statistics Review, 1975-2009 (Vintage 2009
Populations), National Cancer Institute. Bethesda, MD, http://seer.cancer.gov/
csr/1975_2009_pops09/, based on November 2011 SEER data submission, posted
to the SEER web site, April 2012.
2.Brinton LA, Sherman ME, Carreon JD, Anderson WF. Recent trends in breast
cancer among younger women in the United States. J Natl Cancer Inst. 2008 Nov
19;100 (22):1643-8. Epub 2008 Nov 11.
3.Andaya AA, Enewold L, Horner MJ, Jatoi I, Shriver CD, Zhu K. Socioeconomic
disparities and breast cancer hormone receptor status. Cancer Causes Control
2012 23: 951-958.
Surveillance, Epidemiology, and End Results (SEER) Program (www.seer.cancer.gov) SEER*Stat
Database: Mortality - All COD, Aggregated With State, Total U.S. (1969-2009) .
4.Krieger N, Chen JT, Ware JH, Kaddour A. Race/ethnicity and breast cancer
estrogen receptor status: impact of class, missing data, and modeling assumptions.
Cancer Causes Control 2008 19 (10): 1305-1318.
5.Surveillance, Epidemiology, and End Results (SEER) Program (www.seer.cancer.
gov) SEER*Stat Database: Incidence - SEER 18 Regs Research Data + Hurricane
Katrina Impacted Louisiana Cases, Nov 2011 Sub (1973-2009 varying) - Linked
To County Attributes - Total U.S., 1969-2010 Counties, National Cancer Institute,
DCCPS, Surveillance Research Program, Surveillance Systems Branch, released
April 2012, based on the November 2011 submission.
Risk Factors for Ovarian Cancer in African-American Women
Investigator: Michele L. Cote, PhD
25
20
15
10
5
2000
Ovarian cancer survival is poor, with an overall 5-year survival of
43.9% for white women and 38.8% for African-American women.
Although ovarian cancer accounts for 3% of cancer diagnoses
among women, it is the fifth most common cause of cancer mortality accounting for 6% of cancer deaths. The high mortality is largely
attributable to late detection, with 79% of the cancers diagnosed in
regional or distant stages, when there is already spread beyond the
pelvis. (Altekruse, et al, 2010)
Ovarian cancer mortality statistics are even grimmer for AfricanAmerican women. The ratio of mortality to incidence rates is 74%
for African-Americans compared to 65% for whites. (SEER) It has
been noted that five-year survival rates for white women increased
from 36% to 45% from the period 1975-77 to 1996-2004, whereas
5-year survival for African-Americans has decreased, dropping
from 43% in 1975-77 to 38% in 1996-2004. (SEER) Thus, despite
improvements in surgical techniques and chemotherapy 5-year
survival has not improved for African-American women after a
diagnosis of ovarian cancer.
Risk factors for ovarian cancer have been explored primarily in
white women. In the few studies that have included AfricanAmerican women, reproductive factors such as a greater number of
pregnancies, oral contraceptive use and tubal ligation are associated
with reduced risk of ovarian cancer in both races (John 1993, Ness
2000, and Mooreman 2008). These analyses have all been limited
by the small number of African-Americans included in the studies.
Thus, there is a need to further investigate ovarian cancer risk factors among African-American women, including known genetic risk
factors (i.e. BRCA mutations) and other less-common exposures.
Joining eight other institutions and led by investigators at Duke
University, Dr. Michele Cote and researchers at the Metropolitan
Detroit Cancer Surveillance System (MDCSS) of Wayne State University have embarked upon a study of ovarian cancer in AfricanAmerican women. During the course of the 5 year study, investigators plan to interview and obtain blood specimens from 1,000
African-American women with ovarian cancer and 1,000 healthy
women. Medical record release and yearly follow up will also be
requested of the women with ovarian cancer. MDCSS hopes to
contribute about 13% of the study subjects. The goal of the study
is to examine epidemiologic and genetic risk factors for ovarian
cancer among African-American women.
1975
1980
1985
1990
1995
2000
2005
YEAR OF DIAGNOSIS
White
African-American
Age-Adjusted Incidence-based Mortality from
Malignant Ovarian Cancer, 1973 – 2009
25
20
RATE PER 100,000
20
Age-Adjusted Incidence of Malignant
Ovarian Cancer by Race, SEER-9, 1973 – 2009
RATE PER 100,000
Ovarian cancer is diagnosed in more than
22,000 women in the United States each year,
making it the 9th most commonly diagnosed
cancer in women. (Jemal et al, 2009) Incidence
rates are higher among white women than
African-American women (14.1/100,000 and
10.1/100,000, respectively), and incidence
rates have been declining over time. (Altekruse, et al, 2010) The decrease in incidence
trends from 1975 to 2005 is much higher among white women
(-2.7%) than African-Americans (-0.4%). The median age at diagnosis is younger among African-Americans compared to whites (61
versus 64 years). (Altekruse, et al, 2010)
15
10
5
2000
1975
1980
1985
1990
1995
2000
2005
YEAR OF DEATH
References:
1.
2.
3.
4.
5.
Altekruse SF, Kosary CL, Krapcho M, Neyman N, Aminou R, Waldron W, Ruhl J, Howlader N, Tatalovich Z, Cho H, Mariotto A, Eisner MP, Lewis DR, Cronin K, Chen HS, Feuer EJ, Stinchcomb DG, Edwards BK (eds). SEER Cancer Statistics Review, 1975-2007, National Cancer Institute. Bethesda, MD, http://seer.cancer.
gov/csr/1975_2007/, based on November 2009 SEER data submission, posted to the SEER web site, 2010.
Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics, 2009. CA Cancer J Clin. 2009 Jul-Aug;59(4):225-49.
John EM, Whittemore AS, Harris R, Itnyre J Characteristics relating to ovarian cancer risk: collaborative analysis of seven U.S. case-control studies. Epithelial ovarian cancer in black women. Collaborative Ovarian Cancer Group.J Natl Cancer Inst. 1993 Jan 20;85(2):142-7.
Moorman PG, Palmieri RT, Akushevich L, Berchuck A, Schildkraut JM Am J
Epidemiol. Ovarian cancer risk factors in African-American and white women. 2009 Sep 1;170(5):598-606.
Ness RB, Grisso JA, Cottreau C, Klapper J, Vergona R, Wheeler JE, Morgan M, Schlesselman JJ. Factors related to inflammation of the ovarian epithelium and risk of ovarian cancer. Epidemiology. 2000 Mar;11(2):111-7.
Patterns of Care Study
Ikuko Kato (Principal Investigator), PhD, Patricia Arballo-Spong, BS, Ann S. Bankowski, MSW, Kari A. Borden, RHIT, CTR
SEER Patterns of
Care (POC) Studies
have been funded by
the National Cancer
Institute in order to
assess the incorporation of state-of-theart cancer treatments
into clinical practice
and the extent to
which cancer patients
receive such treatments. These studies are designed to describe, characterize, and
compare practice patterns and treatments provided for cancer in
different geographic areas of the United States. The Metropolitan
Detroit Cancer Surveillance System (MDCSS) has participated in
POC studies to provide representative samples from metropolitan
Detroit since 1997, when this study was integrated into the SEER
program. The results of POC studies have been summarized by
NCI staff and disseminated in scientific journals and professional
meetings; and used to develop educational or training opportunities
to improve the use of state-of-the-art cancer therapy in community
practice through collaboration with professional organizations.
Each year three to four types of cancer are selected for this study.
Table 1 lists cancers studied in years 1997 to 2010. These pertain
to patients diagnosed in years 1995 -2008, i.e., 2 years behind
the years of the survey, to allow for sufficient time to capture all
care that patients received during the initial course of treatment.
Choices of cancers reflect the introduction of new treatment methods, therapeutics, diagnostic tests, predictive and prognostic markers for particular cancer sites, as well as special research interests.
Examples include use of less invasive surgeries such a laparoscopic
resection and cryosurgery, radioimmunotherapy, new monoclonal
antibodies and antiangiogenic agents, active surveillance for prostate cancer, and HER2 and Oncotype tests for breast cancer. Some
cancers have been chosen more than once during this period to
examine chronological trends.
For the past 10 years, approximately 340 cases per year have been
included from the MDCSS in this study. These samples are randomly selected by a computer program at a time point when the number
of selected cancer cases reach 95% of the projected number of cases
for a given year. There are two major components in POC studies:
(1) re-abstracting medical records in more detail beyond regular
abstracts required for SEER and (2) physicians’ surveys. The former
includes individual chemo-, immuno-, hormona-therapeutic agents,
in addition to other new treatments/tests/markers of interest.
SEER abstracting staff complete these tasks at all facilities where a
patient received diagnosis and treatments. Typically, multiple medical record abstracts are required per patient. The physicians’ survey
is mailed to a follow-up physician identified through the MDCSS.
This is a very crucial part of the study because private physicians
provide a large proportion of treatment received by cancer patients, particularly adjuvant chemotherapy, hormonal therapy and
immunotherapy, on an outpatient basis, while the current SEER
program is focused primarily on collection of treatment data from
hospitals, radiation oncology facilities and larger clinics. Thus, our
ability to assess the extent to which such therapies are provided, the
characteristics of patients and providers in relation to treatments
rendered and the efficacy of those treatments depends upon the use
of supplemental data collection efforts such as the POC Study.
Table 1 - Cancers selected for SEER
Patterns of Care (POC) Studies, 1997 – 2010
Year of Survey
Cancer Sites/Types
1997
Bladder, Breast, Colorectal, Melanoma
1998
Childhood Cancer, Non-Small Cell Lung, Ovarian, Melanoma
1999
Childhood Brain Stem, DCIS Breast, Cervical, Head and Neck
2000
Chronic Lymphocyctic Leukemia, Pancreas, Prostate, Corpus Uteri
2001
Vulva, Testicular, Non-Hodgkin’s Lymphoma (NHL), Multiple Myeloma
2002
Breast, Cervical, Colorectal
2003
Cervical, Lower Esophageal/Gastric, Melanoma
2004
Ovarian, Prostate, Sarcoma
2005
B-Cell Lymphoma, Bladder, Chronic Myelogenous Leukemia, Multiple Myeloma
2006
Node negative, ER positive female Breast, Male Breast, Head and Neck, Kidney
2007
Breast, Colorectal, Non-Small Cell Lung
2008
Glioblastoma, Thyroid, Cancers in Adolescent and Young Adult
2009
Small Cell Lung, Acute Myeloid Leukemia, Multiple Myeloma, Liver
2010
Prostate, NHL, Chronic Lymphocytic Leukemia, Gastrointestinal Stromal Tumors
Since 2005, we have formed a strong POC study team within the
MDCSS, which includes Patricia Arballo-Spong, SEER Project Coordinator; Ann Bankowski, Research Assistant; Kari Borden, Abstracting Supervisor and Dr. Ikuko Kato as the Principal Investigator.
Through this teamwork we have implemented several improvements in this study; e.g., development of an efficient database to
track the progress of each record and to search alternative follow-up
physicians, and electronic data submission and data security. We
are very proud that we have contributed high quality data to this
SEER-supported study.
Finally, we can not thank enough, all physicians who responded
to a survey over these years despite their extremely busy work
schedules. Through the end of 2009, the POC studies produced 28
publications, including Annual Report to the Nation on the status
of Cancer (1). These publications can be accessed online at http://
healthservices.cancer.gov/surveys/poc/
Reference:
1.
Edwards BK, Brown ML, Wingo PA, Howe HL, Ward E, Ries LA, Schrag D,
Jamison PM, Jemal A, Wu XC, Friedman C, Harlan L, Warren J, Anderson
RN, Pickle LW. Annual report to the nation on the status of cancer, 1975-2002,
featuring population-based trends in cancer treatment. J Natl Cancer Inst 2005
Oct 5;97(19):1407-27.
21
Genetic Pathways of Inflammation and Risk of Lung Cancer
and Chronic Obstructive Pulmonary Disease
Investigator: Ann G. Schwartz, PhD, MPH
22
It has been shown that a family history of COPD increases risk of
lung cancer, even after accounting for familial clustering of smoking. This suggests a common underlying genetic contribution to
these two diseases. The biologic mechanisms linking COPD and
lung cancer are not known, but chronic inflammation of lung tissue
is likely to play a role. Both lung cancer and COPD develop in a lung
environment battling chronic inflammation, which is developed
over a lifetime from cigarette smoke exposure. This environment
is conducive to genetic damage, cancer initiation and progression.
We do not yet know which specific genetic inflammatory mediators
either jointly or independently promote COPD and tumor development. Although biologic responses to tobacco smoke underlie both
COPD and lung cancer, the inflammatory processes leading
to COPD and those associated with lung cancer may be different.
This is suggested by the absence of COPD features in some lung cancer patients and the absence of lung cancer in some COPD patients.
Which inflammation-producing genetic pathways are activated
and/or suppressed is likely to determine the course of disease
development among smokers. However, no comprehensive study
evaluating multiple genetic markers of inflammation in smokers
with lung cancer compared to smokers with COPD and smokers without either disease has ever been conducted. In addition,
African-Americans, who are more likely to develop lung cancer but
are less often diagnosed with COPD than Whites, have historically
not been included in large numbers in studies of genetic variation in
inflammation-related genes. This study, called the “InHALE Study”,
Age-Adjusted Incidence Rates of
Lung & Bronchus Cancers, SEER-9, 1973 – 2009
100
80
RATE PER 100,000
Lung cancer is the leading cause of cancer deaths and chronic obstructive pulmonary disease (COPD) is the fourth leading cause of
all adult deaths in the United States. COPD is a heterogeneous set
of diseases, including emphysema and chronic bronchitis. Lung
cancer and COPD together account for substantial deaths and
morbidity. Five year survival following a lung cancer diagnosis is
only 15%. African-Americans are hardest hit by lung cancer, having
both the highest rates of new lung cancers and lung cancer deaths
than any other racial or ethnic group. Smoking contributes to both
COPD and lung cancer. About 15% of smokers will develop at least
one of these diseases. About 85-90% of all lung cancers and COPD
diagnoses are attributable to cigarette smoking. Having COPD
also contributes to the risk of getting lung cancer. After being
diagnosed with COPD, the risk of subsequently developing lung
cancer increases about 2-fold, even among people who have never
smoked. This risk seems to be most closely associated with a history
of emphysema. While low dose CT has recently been shown to be
an accepted screening method for lung cancer, population screening
is not yet under way. Without screening, lung cancer is identified
in late stages and it is often too late for surgical treatment. Neither
lung cancer, nor COPD are particularly responsive to treatment.
Therefore it is vitally important to understand the underlying
susceptibility to lung cancer and COPD, in order to understand the
carcinogenic process and to identify those persons at highest risk of
getting these diseases. Once we understand who is at greatest risk
of getting lung cancer and COPD, those at risk can be targeted for
screening and prevention interventions.
60
40
20
1975
1980
1985
1990
1995
2000
2000
2005
YEAR OF DIAGNOSIS
White
African-American
Source: Surveillance, Epidemiology, and End Results (SEER) Program (www.seer.cancer.gov) SEER*Stat
Database: Incidence - SEER 9 Regs Research Data, Nov 2010 Sub (1973-2008) <Katrina/Rita Population Adjustment> - Linked To County Attributes - Total U.S., 1969-2009 Counties, National Cancer Institute, DCCPS, Surveillance Research Program, Cancer Statistics Branch, released April 2011 (updated 10/28/2011),
based on the November 2010 submission.
Note: Rates are per 100,000 and age-adjusted to the 2000 US Std Population (19 age groups - Census P251130) standard.
will investigate the hypotheses that genetic variation in inflammation-related genes contributes to the development of lung cancer
and that this association varies by the presence or absence of COPD,
and by race.
To study the underlying inflammatory mechanisms linking COPD
and lung cancer, we will evaluate single nucleotide polymorphisms
(SNPs) in inflammatory pathway genes. Our goal is to develop a
genetic profile that predicts susceptibility to lung cancer with and
without COPD in response to tobacco exposure. Our study will expand on previous work by incorporating COPD phenotyping using
CT diagnosis of emphysema and spirometry to evaluate lung function. There has not been a study of these pathway genes in AfricanAmericans, a group that is less likely to report COPD, smokes fewer
cigarettes, but sadly, is more likely to be diagnosed and die of lung
cancer than Whites. We will invite patients from two large, urban
health systems, Karmanos Cancer Institute (KCI) and Henry Ford
Health System (HFHS), to participate in this important study. We
will recruit 2050 lung cancer cases and 2050 smokers with and
without COPD. We hope to equally include African-Americans and
Whites in this study, so we can better understand why AfricanAmericans are harder hit with lung cancer and design better prevention strategies for everyone. All study participants will be invited to
complete a risk factor questionnaire, undergo CTs and spirometry,
provide a blood and saliva sample, and when available a tissue
block, so that we can investigate the genetic variation contributing
to inflammation and thereby potentially identify a way to prevent
or reduce inflammation that may contribute to development of
COPD and lung cancer. No other large study has been done that
includes detailed phenotyping of lung cancer and COPD and jointly
evaluates inflammatory genes in germline DNA and target tissue
in the same individuals. This work will lead to a better understanding of the inflammatory process in lung carcinogenesis, provide
avenues for the identification of a high risk group for intervention,
and provide insight into possible treatment options.
InHALE, as well as all studies conducted through the MDCSS,
follows strict confidentiality guidelines during all aspects of the
research study. Independent Institutional Review Boards (IRB) that
protect human rights during research oversee the study, both at
KCI (overseen by the Wayne State University IRB) and Henry Ford
Health System. Each participant will be given a consent form that
describes all aspects of the study and who they can contact with any
questions. In this way, each participant is fully informed about the
study and how the study will be conducted, before deciding if they
wish to participate. All confidential data is maintained in locked
files and there is only limited access to computerized data. Paper
records that contain identifiers are kept separate from the risk
factor questionnaires. The risk factor questionnaires are identified only by a unique ID number, to hide the study participant’s
identity and to keep the information they provide confidential.
The interview will include questions about health history including
history of other lung diseases and inflammatory conditions, use of
anti-inflammatory drugs, such as aspirin, smoking history, passive
smoking history, height, weight, occupation, and exposure history.
Blood and other biological samples will also be labeled with only a
unique ID number.
This 5 year study into the genetic inflammatory pathways underlying development of COPD and lung cancer is now enrolling patients
at both the Karmanos Cancer Institute and the Henry Ford Health
Gene content for the custom,
focused SNP array (640 genes total, ~ 6700 SNPS).
(243 genes)
Lung Cancer
(84 genes)
References:
1. SEER Cancer Statistics Review, 1975-2006. 2009. http://SEER.cancer.gov/csr.
Accessed June 1, 2009.
2. Mathers CD, Loncar D. Projections of global mortality and burden of disease from
2002 to 2030. PLoS Med. Nov 2006;3(11):e442. PMID: 17132052
3 Sethi JM, Rochester CL. Smoking and chronic obstructive pulmonary disease.
Clinics in chest medicine. Mar 2000;21(1):67-86, viii. PMID: 10763090
4. CDC. Cigarette smoking among adults--United States, 2002. Morbidity and
Mortality Weekly Report. 2004;53(20):428-431. PMID:
5.
Mannino DM, Aguayo SM, Petty TL, Redd SC. Low lung function and incident
lung cancer in the United States: data From the First National Health and
Nutrition Examination Survey follow-up. Archives of internal medicine. Jun 23
2003;163(12):1475-1480. PMID: 12824098
6. Schabath MB, Delclos GL, Martynowicz MM, et al. Opposing effects of
emphysema, hay fever, and select genetic variants on lung cancer risk. Am J
Epidemiol. Mar 1 2005;161(5):412-422. PMID: 15718477
7. Littman AJ, Thornquist MD, White E, Jackson LA, Goodman GE, Vaughan TL.
Prior lung disease and risk of lung cancer in a large prospective study. Cancer
Causes Control. Oct 2004;15(8):819-827. PMID: 15456995
8.
Turner MC, Chen Y, Krewski D, Calle EE, Thun MJ. Chronic obstructive
pulmonary disease is associated with lung cancer mortality in a prospective study
of never smokers. American journal of respiratory and critical care medicine.
Aug 1 2007;176(3):285-290. PMID: 17478615
9. Mayne ST, Buenconsejo J, Janerich DT. Familial cancer history and lung cancer risk in United States nonsmoking men and women. Cancer Epidemiol Biomarkers Prev. Dec 1999;8(12):1065-1069. PMID: 10613338
Inflammation
COPD
System. Should your physician invite you to participate in this
important research, please consider saying “Yes.” The physician will
then put you in contact with the study interviewers. They will be
available during normal clinic appointments to meet with you. This
study will also enroll people who do not have lung cancer, but who do
smoke, for comparison purposes. You may see our study brochures
handed out at your church, area health fairs or cancer awareness
programs; you may also hear or see ads for the “InHALE Study”.
Together, we hope to prevent future lung cancers and COPD.
(358 genes)
10. Wu AH, Yu MC, Thomas DC, Pike MC, Henderson BE. Personal and family history of lung disease as risk factors for adenocarcinoma of the lung. Cancer Res. Dec 15 1988;48(24 Pt 1):7279-7284. PMID: 3191498
11. Schwartz AG, Ruckdeschel JC. Familial lung cancer: genetic susceptibility and relationship to chronic obstructive pulmonary disease. American journal of
respiratory and critical care medicine. Jan 1 2006;173(1):16-22. PMID: 16141445
12. Gohagan J, Marcus P, Fagerstrom R, Pinsky P, Kramer B, Prorok P. Baseline findings
of a randomized feasibility trial of lung cancer screening with spiral CT scan vs
chest radiograph: the Lung Screening Study of the National Cancer Institute. Chest.
Jul 2004;126(1):114-121. PMID: 15249451
23
The Epidemiology Research Core
Director: Kendra Schwartz, MD, MSPH; Co-Director: Fawn D.Vigneau, JD, MPH;
Project Coordinator: Patricia Arballo-Spong; Administrator: Sharon Moton
Wayne State University
(WSU) and Karmanos Cancer
Institute (KCI) house the
Metropolitan Detroit Cancer
Surveillance System (MDCSS)
and the Epidemiology
Research Core (ERC). The
MDCSS is a National Cancer
Institute (NCI)-funded,
population-based Surveillance Epidemiology and End Results
(SEER) cancer registry. The MDCSS collects cancer data for the
metropolitan Detroit area (Wayne, Oakland and Macomb counties)
population of nearly four million residents. Data are collected from
approximately 60 different hospitals, clinics, pathology laboratories
and radiation therapy facilities in the area (Figure 1).
data for population-based investigations of cancer prevention,
etiology, treatment and outcomes. Specifically, the Epidemiology
Research Core provides:
• Consultation. Consultation and collaborative research expertise focused
on study design, proposal development, IRB applications, and interpretation
of population-based local and national SEER data.
• Rapid Case Ascertainment. Rapid identification and ascertainment of
eligible study participants, requiring review of all pathology reports
indicating a cancer diagnosis at approximately 60 metropolitan area
hospitals, clinics, pathology laboratories and radiation therapy facilities
and rapid collection of patient demographic information. This speeds up
case identification for population-based studies, with most cases identified
within one to two months of diagnosis.
• Control Identification. Identification of population-based control groups
for case-control study designs via random digit dialing, Centers for Medicaid
and Medicare Services (CMS) records, Department of Motor Vehicles (DMV)
records, and other mechanisms.
• Research Support and Training. Liaison for WSU IRB, regulatory
training of epidemiology study staff, oversight and research support for
epidemiology studies.
24
• Collection and Abstraction of Medical Records. Collection of medical
records and abstracting of study-specific data which are not available in the
MDCSS registry.
• Biospecimen Collection. Collection of biologic specimens, including
blood, buccal cells and tumor blocks, from study participants and/or
diagnosing hospitals.
• Database Query. Response to requests for population-based data
requiring query of the MDCSS and/or the national SEER Limited-Use data
files for descriptive analyses.
Figure 1. MDCSS/Detroit SEER – Participating Facilities
The MDCSS receives SEER contractual funding for the conduct of
cancer case-finding, medical record abstraction for SEER-required
information, vital status follow-up, and the provision of data back
to the contributing facilities for clinical and accreditation purposes.
NCI contractual funding for the MDCSS covers only basic registry
operations. The ERC was developed to support population-based
cancer research. The services of the ERC represent populationbased research activities that are outside the scope of the MDCSS/
SEER cancer registry data collection. The ERC does this by
accessing metropolitan Detroit cancer case data for research,
epidemiology consulting and collaboration with researchers
conducting investigations in cancer prevention, etiology, treatment
and outcomes. The ERC benefits cancer research in metropolitan
Detroit by centralizing access to SEER data and standardizing
patient, physician and hospital interactions for population-based
research purposes.
SEER data are complex and require knowledge of and expertise
with the data management system for manipulation. Furthermore,
strict confidentiality measures must be maintained. The ERC
maintains patient and provider confidentiality by performing
all patient, physician and hospital contact for the conduct of
population-based research in-house. ERC faculty and staff have
extensive expertise in using both local and national SEER data and
provide the necessary support for access and utilization of these
• Database Linkage. Linkage of outside data sources to MDCSS data for the
collection of patient demographics, treatment and survival data.
Current projects utilizing ERC services include:
• Inflammation Pathways and COPD in the Development
of Lung Cancer
• Life Course Energy Balance and Breast Cancer Risk in Black/
White Women Under 50
• Patient and Provider Influences on Disparities in Colorectal
Cancer Care
• The Epidemiology of Ovarian Cancer in African-American Women
Researchers wishing to utilize Metropolitan Detroit SEER data can contact:
Kendra L. Schwartz, MD, MSPH
Director, Epidemiology Research Core
[email protected]
Fawn D. Vigneau, JD, MPH
Co-Director, Epidemiology Research Core
[email protected]
Prices for use of ERC services are listed on our web-site:
https://research1.karmanos.org/research/Home2/Researchers2/CoreFacilities/
EpidemiologyResearch/ServicesFees.aspx
List of Studies Conducted at the
Metropolitan Detroit Cancer Surveillance System
The Metropolitan Detroit Cancer Surveillance System participates in
a number of collaborative research projects. A brief description of a
sample of these projects conducted during the past 2 years follows.
foundation to conduct studies of other cancers in African-American
women in the future. We will expand the examination of genetic
association to a more comprehensive genome-wide association
study once data collection is complete.
Inflammation Pathways and COPD in the
Development of Lung Cancer
Dr. Cote commenced rapid case ascertainment in December 2010
for the Metropolitan Detroit site of the multi-site study. The
national collaboration will attempt to enroll 1,000 cases and 1,000
controls at nine sites across the U.S. The Detroit site started
Biospecimen Collection in October, 2011 and will collect blood and
tissue on 35 cases annually.
A. Schwartz
The biologic mechanisms linking COPD and lung cancer are
unknown, but chronic inflammation is likely to play a role. This
study will evaluate SNPs and copy number variation in genes
in inflammatory pathways. The study will expand on previous
work by incorporating COPD phenotyping using CT diagnosis of
emphysema and pulmonary function testing (PFT). It also will
expand on the panel of genes beyond those few already evaluated,
will incorporate copy number variation as well as non-synonymous
and functional SNPs, and will include a large African-American
population. There has not been a study of these pathway genes
in African-Americans, a group that is less likely to report COPD,
smokes fewer cigarettes, but is more likely to be diagnosed with
lung cancer than whites. It is hypothesized that genetic variation
in inflammation-related genes contributes to the development of
lung cancer and this association varies by the presence or absence
of COPD, and by race. The goal is to develop a genetic profile based
on SNPs and copy number variation in inflammation pathway genes
that predicts susceptibility to lung cancer with and without COPD
in response to tobacco exposure.
Transdisciplinary Research in Cancer
of the Lung (TRICL)
A. Schwartz/C. Amos
This study integrates data from multiple genome-wide association
studies in lung cancer and carries these findings forward to
characterize mechanisms by which the identified genes influence
lung cancer risk. We will use results from extended epidemiological
studies to develop refined risk models to predict lung cancer risk
and to validate the results using data from cohort studies.
The Epidemiology of Ovarian Cancer in
African-American Women
A Genome Wide Study of Lung Cancer
in Never Smokers
A. Schwartz/O. Gorlova
This study will identify genetic architecture of the predisposition
to lung cancer in never smokers.
25
Genetic Susceptibility for Lung Cancer in
African-Americans
A. Schwartz/C. Amos
The aim of this work is to validate novel genetic variants identified
in genome-wide association studies in African-American lung
cancer cases and age, race and gender-matched controls identified
from our existing study populations.
Pancreatic Cancer Genetic Epidemiology Consortium
A. Schwartz/G. Petersen
This project establishes the PACGENE Consortium of six
institutions throughout the Unites States and Canada whose
purpose is to identify and recruit kindreds with familial pancreatic
cancer for gene discovery research. Family history and risk factor
information will be collected from participating family members,
along with DNA specimens for a 10-cM genome screen for linkage
and subsequent fine mapping of selected chromosomal regions. Dr.
Schwartz is the PI of a subcontract to the Mayo Clinic, where the PI
of the Consortium is Dr. Gloria Petersen.
A. Schwartz/M. Cote
To address the lack of knowledge regarding ovarian cancer among
African-American women, investigators at nine institutions in
areas with relatively large numbers of African-Americans in the
population (North Carolina, South Carolina, Georgia, Alabama,
New Orleans, New Jersey, Ohio, Illinois, and Detroit) will conduct
a population-based, case-control study to assess etiologic risk
factors for African-Americans. Further, we will obtain treatment
and outcome information to evaluate prognostic factors in
ovarian cancer cases. We anticipate that this study will provide a
Genetic Epidemiology of Lung Cancer I
A. Schwartz
This study is designed to create a family registry and search
for lung cancer genes in high risk lung cancer families.
A supplemental study will identify additional lung cancer cases
with family histories for a future GWAS.
List of Studies Conducted at the
Metropolitan Detroit Cancer Surveillance System (cont.)
Genetic Epidemiology of Lung Cancer
A. Schwartz
This study is a case-control study of lung cancer in AfricanAmericans that will use admixture mapping to identify genes for
lung cancer. This grant has been continuously funded since 1999
and also focused on familial aggregation and candidate gene studies
in early onset and never smoking lung cancer cases.
A supplement is focused on reconsenting participants for use of
their biospecimens in an NCI-directed GWAS in African-Americans.
Life Course Energy Balance and Breast Cancer Risk
in Black/White Women Under 50
K. Schwartz/E. Velie
26
Breast cancer in younger women (under age 50) is poorly
understood and understudied. Moreover, it is associated with a
worse prognosis than breast cancer in older women. Potentially
modifiable risk factors related to energy balance over the life course
(e.g. growth and puberty patterns, body mass, insulin resistance,
diet and physical activity) have been implicated in the etiology of
breast cancer in younger women. The overall objective of this study
is to investigate, in a socioeconomically diverse population of Black
and White women, whether life course energy balance pathways are
associated with breast cancer risk. The study will also investigate
the association of energy balance with the different breast cancer
molecular subtypes. Dr. Kendra Schwartz is the local PI for this
National Cancer Institute funded study.
Racial, Ethnic and SES Disparities in Quality
of Breast Cancer Systemic Therapy
K. Schwartz/J. Griggs
The study will conduct a medical record review and tests on
previously removed breast cancer tumor tissue to explore the
quality of adjuvant chemotherapy and hormonal therapy as well as
variations based on race and ethnicity. It is a companion study to
“Racial/Ethnic Disparities in Work and QOL Outcomes in Survivors
of Breast Cancer” for patients who were initially diagnosed with
invasive breast cancer without distant metastasis.
Racial/Ethnic Disparities in Work and QOL Outcomes
in Survivors of Breast Cancer
K. Schwartz/S. Hawley
The study will re-survey female breast cancer patients four years
after diagnosis to explore employment changes, quality of life
ratings, decisions about adjuvant therapy, and variations in
experience based on race and ethnicity. This is a follow-up study
designed specifically for patients who enrolled in “Health System
Factors and Patient Outcomes in Breast Cancer.”
Disparity in Quality of Breast Cancer Treatment
and Effects on Outcome
K. Schwartz/J. Griggs
In this research study we will investigate the relationship between
the quality of breast cancer adjuvant treatment and disease-free
and overall survival; determine whether racial, socioeconomic,
and weight-based disparities exist in the quality of breast cancer
adjuvant treatment and timing of treatments; and verify that
disparities in outcomes exist.
Health System Factors and Patient Outcomes
in Breast Cancer
K. Schwartz/S. Katz
This study measures the impact of the breast cancer experience
on women, addressing topics such as barriers to diagnosis and
treatment, coordination of care and employment difficulties
related to treatment.
Prostate Cancer Treatment Decision Making ACS Mentored Research Award
K. Schwartz/J. Xu
This study uses both qualitative and quantitative methods to
better understand racial/ethnic disparities in prostate cancer
decision treatment. A structured interview will be conducted with
recently diagnosed prostate cancer patients, prior to treatment.
This information will be used in creating a culturally sensitive
comprehensive instrument to determine factors associated with
prostate cancer treatment choice, which will then be pilot tested as
a mailed questionnaire to recently diagnosed prostate cancer cases
identified through SEER.
Patient and Provider Influences on Disparities in
Colorectal Cancer Care
I. Kato
The investigators seek to examine whether racial disparities in use
of chemotherapy for stage III colorectal cancer are primarily due
to socioeconomic, psychosocial and patient-provider relationship
factors. The project will be conducted in collaboration with the
Atlanta SEER registry under leadership of Dr. Morris at the
University of Michigan.
Analysis of Cancer Rates and Trends Using Census
Tract Variables from Multiple Registries
Oxidative Stress, Antioxidants and Racial
Disparities in Prostate Cancer
I. Kato
C. Bock
Despite the growing recognition of the magnitude and
persistence of health disparities, few indicators for socioeconomic
status (SES) are available in most of the US public health
surveillance databases to date. Using the data from four SEER
registries, Detroit, Hawaii, Utah and Seattle-Puget, that have a
long history of high quality cancer registration and excellent
census tract coverage, we intend to develop common objective and
transferrable census tract SES variables across multi-decennial
data from 1970 to 2000 and examine long-term chronological
trends in cancer survival for selected cancer sites according to
those census tract variables. This investigation may help identify
specific SES groups in the community which require more intensive
intervention in reducing disparities.
This study will examine three genes in the oxidative stress pathway
along with intake of three antioxidants to determine whether
variation in the genes modifies prostate cancer risk associated with
intake of each of the antioxidants.
SEER Patterns of Care Study
I. Kato
The SEER Patterns of Care (POC) Study annually examines
treatment patterns in the U.S. for rotating cancer sites.
The 2012 POC study will collect data on metastatic melanoma,
mesothelioma, oligodendroglioma/astrocytoma, neuroblastoma,
and ovarian cancer diagnosed in the year 2011.
Exploring Common Linkage Regions in
Lung Cancer and COPD
M. Cote
The overall goal of this research plan is to investigate the
importance of chromosome 6 on lung cancer and COPD
development.
A Genome Wide Admixture Scan of Multiple
Myeloma in African-Americans
J. Zonder/C. Bock
African-Americans experience the highest risk of multiple myeloma
in the world. The reason for the higher risk is not known, but there
is evidence that genetic factors might contribute. The purpose of
this research study is to identify the causes of multiple myeloma
in African-Americans, especially genetic risk factors, which we
hope, will lead to better treatments and prevention. We expect
that about 2,000 patients will participate in this study. Patients are
being recruited from California, Alabama, New Jersey, Louisiana,
Detroit, Atlanta, Houston, Baltimore and Chicago. This study is
being led by Wendy Cozen, DO, MPH.
Genetic Susceptibility to Cancers of the Prostate
and Urinary Bladder
J. Beebe-Dimmer
The overall goal of this research is to estimate the effects of
genetic factors on risk of prostate and bladder cancers in two
established studies.
27
Metropolitan Detroit Cancer
Surveillance System/SEER Publications 2010-2012
In Press
Becker N, Falster MO, Vajdic CM, de Sanjose S, Martinez-Maza O, Bracci
PM, Melbye M, Smedby KE, Engels EA, Turner J, Vineis P, Costantini
AS, Holly EA, Spinelli JJ, La Vecchia C, Zheng T, Chiu BCH, Montella M,
Cocco P, Maynadie M, Foretova L, Staines A, Brennan P, Davis S, Severson
R, Cerhan JR, Breen EC, Birmann B, Cozen W, Grulich AE, Newton
R. Self-reported history of infections and the risk of non-hodgkin
lymphoma: an InterLymph pooled analysis. Int J Cancer. In press.
Keegan TH, Lichtensztajn DY, Kato I, Kent EE, Wu XC, West MM, Hamilton
AS, Zebrack B, Bellizzi KM, Smith AW, and the AYAHSCG. Unmet
adolescent and young adult cancer survivors information and service
needs: a population-based cancer registry study. J Cancer Surviv. In press.
Parsons HM, Harlan LC, Lynch CF, Hamilton AS, Wu XC, Kato I, Schwartz
SM, Smith AW, Keel G, Keegan TH. Impact of Cancer on Work and
Education Among Adolescent and Young Adult Cancer Survivors. J Clin
Oncol. In press.
Schwartz AG, Schwartz K, Harris J. USA, Michigan, Metropolitan Detroit.
In Cancer Incidence in Five Continents, Volume IX: IARC Scientific
Publication. In press.
28
Simon MS, Lamerato L, Krajenta R, Booza JR, Ruterbusch JJ, Kunz S,
Schwartz K. Racial Differences in the Use of Adjuvant Chemotherapy for
Breast Cancer in a Large Urban Integrated Health System. International
Journal of Breast Cancer. In press.
Wang SS, Hartge P, Cerhan J, Cozen W, Davis S, Severson R, Chatterjee N,
Yeager M, Chanock S, Rothman N. Immune gene polymorphisms and risk
of non-Hodgkin lymphoma in the NCI-SEER case control study. J Clin
Oncol. In press.
Wang SS, Purdue M, Cerhan JR, Zheng T, Menashe I, Armstrong B, Lan Q,
Hartge P, Kricker A, Zhang Y, Morton LM, Valdjic C, Holford T, Severson
RK, Grulich A, Leaderer B, Davis S, Zahm S, Cozen W, Yeager M, Chanock
SJ, Chatterjee N, Rothman N. Common Genetic Variations in the Tumor
Necrosis Factor (TNF) and TNF Receptor Superfamilies and Nuclear
Factor Kappa Beta Transcription Factors and Risk of Non-Hodgkin
Lymphoma. PLoS ONE. In press.
Wang S, Levin A, Smolinski S, Vigneau F, Levin N, Tainsky M. Breast
Cancer and Other Neoplasia in Women with Neurofibromatosis Type 1:
A Retrospective Review of Cases in the Detroit Metropolitan Area. Am J
Med Genet Part A. In press.
2012
Al-Sukhni W, Joe S, Lionel AC, Zwingerman N, Zogopoulos G, Marshall
CR, Borgida A, Holter S, Gropper A, Moore S, Bondy M, Klein AP,
Petersen GM, Rabe KG, Schwartz AG, Syngal S, Scherer SW, Gallinger
S. Identification of germline genomic copy number variation in familial
pancreatic cancer. Hum Genet. 2012. [Epub ahead of print]
Beebe-Dimmer JL, Cetin K, Shahinian V, Morgenstern H, Yee C, Schwartz
KL, Acquavella J. Timing of androgen deprivation therapy use and
fracture risk among elderly men with prostate cancer in the United
States. Pharmacoepidemiol Drug Saf. 2012;21(1):70-8.
Bellizzi KM, Smith A, Schmidt S, Keegan TH, Zebrack B, Lynch CF, Deapen
D, Shnorhavorian M, Tompkins BJ, Simon M, and the A, Young Adult
Health O, Patient Experience Study Collaborative G. Positive and negative
psychosocial impact of being diagnosed with cancer as an adolescent or
young adult. Cancer. 2012. [Epub ahead of print]
Boleij A, Roelofs R, Danne C, Bellais S, Dramsi S, Kato I, Tjalsma H.
Selective Antibody Response to Streptococcus gallolyticus Pilus Proteins
in Colorectal Cancer Patients. Cancer Prev Res (Phila). 2012;5(2):260-5.
Chen LS, Saccone NL, Culverhouse RC, Bracci PM, Chen CH, Dueker N, Han
Y, Huang H, Jin G, Kohno T, Ma JZ, Przybeck TR, Sanders AR, Smith
JA, Sung YJ, Wenzlaff AS, Wu C, Yoon D, Chen YT, Cheng YC, Cho YS,
David SP, Duan J, Eaton CB, Furberg H, Goate AM, Gu D, Hansen HM,
Hartz S, Hu Z, Kim YJ, Kittner SJ, Levinson DF, Mosley TH, Payne TJ,
Rao DC, Rice JP, Rice TK, Schwantes-An TH, Shete SS, Shi J, Spitz MR,
Sun YV, Tsai FJ, Wang JC, Wrensch MR, Xian H, Gejman PV, He J, Hunt
SC, Kardia SL, Li MD, Lin D, Mitchell BD, Park T, Schwartz AG, Shen H,
Wiencke JK, Wu JY, Yokota J, Amos CI, Bierut LJ. Smoking and genetic
risk variation across populations of European, Asian, and AfricanAmerican ancestry--a meta-analysis of chromosome 15q25. Genet
Epidemiol. 2012;36(4):340-51. PMC3387741.
Cote ML, Kam A, Chang CY, Raskin L, Reding KW, Cho KR, Gruber SB,
Ali-Fehmi R. A pilot study of microsatellite instability and endometrial
cancer survival in white and African-American women. Int J Gynecol
Pathol. 2012;31(1):66-72.
Cote ML, Liu M, Bonassi S, Neri M, Schwartz AG, Christiani DC, Spitz
MR, Muscat JE, Rennert G, Aben KK, Andrew AS, Bencko V, Bickeboller
H, Boffetta P, Brennan P, Brenner H, Duell EJ, Fabianova E, Field JK,
Foretova L, Friis S, Harris CC, Holcatova I, Hong YC, Isla D, Janout V,
Kiemeney LA, Kiyohara C, Lan Q, Lazarus P, Lissowska J, Le Marchand
L, Mates D, Matsuo K, Mayordomo JI, McLaughlin JR, Morgenstern H,
Mueller H, Orlow I, Park BJ, Pinchev M, Raji OY, Rennert HS, Rudnai P,
Seow A, Stucker I, Szeszenia-Dabrowska N, Dawn Teare M, Tjonnelan A,
Ugolini D, van der Heijden HF, Wichmann E, Wiencke JK, Woll PJ, Yang
P, Zaridze D, Zhang ZF, Etzel CJ, Hung RJ. Increased risk of lung cancer
in individuals with a family history of the disease: A pooled analysis from
the International Lung Cancer Consortium. Eur J Cancer. 2012. [Epub
ahead of print]
Cote ML, Colt JS, Schwartz KL, Wacholder S, Ruterbusch JJ, Davis F,
Purdue M, Graubard BI, Chow WH. Cigarette smoking and renal cell
carcinoma risk among black and white Americans: effect modification
by hypertension and obesity. Cancer Epidemiol Biomarkers Prev.
2012;21(5):770-9. PMC3348421.
Djuric Z, Severson RK, Kato I. Association of dietary quercetin with
reduced risk of proximal colon cancer. Nutr Cancer. 2012;64(3):351-60.
PMC3329181.
Jacobs KB, Yeager M, Zhou W, Wacholder S, Wang Z, Rodriguez-Santiago B,
Hutchinson A, Deng X, Liu C, Horner MJ, Cullen M, Epstein CG, Burdett
L, Dean MC, Chatterjee N, Sampson J, Chung CC, Kovaks J, Gapstur SM,
Stevens VL, Teras LT, Gaudet MM, Albanes D, Weinstein SJ, Virtamo J,
Taylor PR, Freedman ND, Abnet CC, Goldstein AM, Hu N, Yu K, Yuan
JM, Liao L, Ding T, Qiao YL, Gao YT, Koh WP, Xiang YB, Tang ZZ, Fan
JH, Aldrich MC, Amos C, Blot WJ, Bock CH, Gillanders EM, Harris CC,
Haiman CA, Henderson BE, Kolonel LN, Le Marchand L, McNeill LH,
Rybicki BA, Schwartz AG, Signorello LB, Spitz MR, Wiencke JK, Wrensch
M, Wu X, Zanetti KA, Ziegler RG, Figueroa JD, Garcia-Closas M, Malats
N, Marenne G, Prokunina-Olsson L, Baris D, Schwenn M, Johnson A,
Landi MT, Goldin L, Consonni D, Bertazzi PA, Rotunno M, Rajaraman
P, Andersson U, Freeman LE, Berg CD, Buring JE, Butler MA, Carreon T,
Feychting M, Ahlbom A, Gaziano JM, Giles GG, Hallmans G, Hankinson
SE, Hartge P, Henriksson R, Inskip PD, Johansen C, Landgren A, McKeanCowdin R, Michaud DS, Melin BS, Peters U, Ruder AM, Sesso HD, Severi
G, Shu XO, Visvanathan K, White E, Wolk A, Zeleniuch-Jacquotte A,
Zheng W, Silverman DT, Kogevinas M, Gonzalez JR, Villa O, Li D, Duell
EJ, Risch HA, Olson SH, Kooperberg C, Wolpin BM, Jiao L, Hassan M,
Wheeler W, Arslan AA, Bueno-de-Mesquita HB, Fuchs CS, Gallinger
S, Gross MD, Holly EA, Klein AP, Lacroix A, Mandelson MT, Petersen
G, Boutron-Ruault MC, Bracci PM, Canzian F, Chang K, Cotterchio M,
Giovannucci EL, Goggins M, Bolton JA, Jenab M, Khaw KT, Krogh
V, Kurtz RC, McWilliams RR, Mendelsohn JB, Rabe KG, Riboli E,
Tjonneland A, Tobias GS, Trichopoulos D, Elena JW, Yu H, Amundadottir
L, Stolzenberg-Solomon RZ, Kraft P, Schumacher F, Stram D, Savage
SA, Mirabello L, Andrulis IL, Wunder JS, Garcia AP, Sierrasesumaga L,
Barkauskas DA, Gorlick RG, Purdue M, Chow WH, Moore LE, Schwartz
KL, Davis FG, Hsing AW, Berndt SI, Black A, Wentzensen N, Brinton LA,
Lissowska J, Peplonska B, McGlynn KA, Cook MB, Graubard BI, Kratz CP,
Greene MH, Erickson RL, Hunter DJ, Thomas G, Hoover RN, Real FX,
Fraumeni JF, Jr., Caporaso NE, Tucker M, Rothman N, Perez-Jurado LA,
Chanock SJ. Detectable clonal mosaicism and its relationship to aging
and cancer. Nat Genet. 2012;44(6):651-8. PMC3372921.
Mina N, Soubani AO, Cote ML, Suwan T, Wenzlaff AS, Jhajhria S, Samarah
H, Schwartz AG. The relationship between chronic obstructive pulmonary
disease and lung cancer in African-American patients. Clin Lung Cancer.
2012;13(2):149-56.
Patel MK, Cote ML, Ali-Fehmi R, Buekers T, Munkarah AR, Elshaikh MA.
Trends in the utilization of adjuvant vaginal cuff brachytherapy and/or
external beam radiation treatment in stage I and II endometrial cancer: a
surveillance, epidemiology, and end-results study. Int J Radiat Oncol Biol
Phys. 2012;83(1):178-84.
Roberts NJ, Jiao Y, Yu J, Kopelovich L, Petersen GM, Bondy ML, Gallinger
S, Schwartz AG, Syngal S, Cote ML, Axilbund J, Schulick R, Ali SZ,
Eshleman JR, Velculescu VE, Goggins M, Vogelstein B, Papadopoulos
N, Hruban RH, Kinzler KW, Klein AP. ATM mutations in patients with
hereditary pancreatic cancer. Cancer Discov. 2012;2(1):41-6.
Rosenberger A, Bickeboller H, McCormack V, Brenner DR, Duell EJ,
Tjonneland A, Friis S, Muscat JE, Yang P, Wichmann HE, Heinrich J,
Szeszenia-Dabrowska N, Lissowska J, Zaridze D, Rudnai P, Fabianova
E, Janout V, Bencko V, Brennan P, Mates D, Schwartz AG, Cote ML,
Zhang ZF, Morgenstern H, Oh SS, Field JK, Raji O, McLaughlin JR,
Wiencke J, LeMarchand L, Neri M, Bonassi S, Andrew AS, Lan Q, Hu W,
Orlow I, Park BJ, Boffetta P, Hung RJ. Asthma and lung cancer risk: a
systematic investigation by the International Lung Cancer Consortium.
Carcinogenesis. 2012;33(3):587-97. PMC3291861.
Schwartz AG. Genetic epidemiology of cigarette smoke-induced lung
disease. Proc Am Thorac Soc. 2012;9(2):22-6. PMC3359114.
Semaan A, Ali-Fehmi R, Munkarah AR, Bandyopadhyay S, Morris RT, Rizk
S, Mert I, Ruterbusch JJ, Cote ML. Clinical/pathologic features and
patient outcome in early onset endometrial carcinoma: A population
based analysis and an institutional perspective from the Detroit
metropolitan area, Michigan. Gynecol Oncol. 2012;124(2):265-9.
Sethi S, Ali-Fehmi R, Franceschi S, Struijk L, van Doorn LJ, Quint W,
Albashiti B, Ibrahim M, Kato I. Characteristics and survival of head and
neck cancer by HPV status: a cancer registry-based study. Int J Cancer.
2012;131(5):1179-86. PMC3319485.
Seward S, Ali-Fehmi R, Munkarah AR, Semaan A, Al-Wahab ZR, Elshaikh
MA, Cote ML, Morris RT, Bandyopadhyay S. Outcomes of patients with
uterine serous carcinoma using the revised FIGO staging system. Int J
Gynecol Cancer. 2012;22(3):452-6.
Wu X, Scelo G, Purdue MP, Rothman N, Johansson M, Ye Y, Wang Z,
Zelenika D, Moore LE, Wood CG, Prokhortchouk E, Gaborieau V,
Jacobs KB, Chow WH, Toro JR, Zaridze D, Lin J, Lubinski J, Trubicka
J, Szeszenia-Dabrowska N, Lissowska J, Rudnai P, Fabianova E, Mates
D, Jinga V, Bencko V, Slamova A, Holcatova I, Navratilova M, Janout V,
Boffetta P, Colt JS, Davis FG, Schwartz KL, Banks RE, Selby PJ, Harnden
P, Berg CD, Hsing AW, Grubb RL, 3rd, Boeing H, Vineis P, Clavel-Chapelon
F, Palli D, Tumino R, Krogh V, Panico S, Duell EJ, Quiros JR, Sanchez
MJ, Navarro C, Ardanaz E, Dorronsoro M, Khaw KT, Allen NE, Buenode-Mesquita HB, Peeters PH, Trichopoulos D, Linseisen J, Ljungberg
B, Overvad K, Tjonneland A, Romieu I, Riboli E, Stevens VL, Thun MJ,
Diver WR, Gapstur SM, Pharoah PD, Easton DF, Albanes D, Virtamo J,
Vatten L, Hveem K, Fletcher T, Koppova K, Cussenot O, Cancel-Tassin G,
Benhamou S, Hildebrandt MA, Pu X, Foglio M, Lechner D, Hutchinson
A, Yeager M, Fraumeni JF, Jr., Lathrop M, Skryabin KG, McKay JD, Gu
J, Brennan P, Chanock SJ. A genome-wide association study identifies a
novel susceptibility locus for renal cell carcinoma on 12p11.23. Hum Mol
Genet. 2012;21(2):456-62. PMC3276284.
2011
Alderman AK, Hawley ST, Morrow M, Salem B, Hamilton A, Graff JJ,
Katz S. Receipt of delayed breast reconstruction after mastectomy:
do women revisit the decision? Ann Surg Oncol. 2011;18(6):1748-56.
PMC3174852.
Beebe-Dimmer J, Morgenstern H, Cetin K, Yee C, Bartoces M, Shahinian V,
Fryzek J, Acquavella J, Schwartz KL. Androgen deprivation therapy and
cataract incidence among elderly prostate cancer patients in the United
States. Ann Epidemiol. 2011;21(3):156-63.
Boffetta P, Jayaprakash V, Yang P, Asomaning K, Muscat JE, Schwartz AG,
Zhang ZF, Le Marchand L, Cote ML, Stoddard SM, Morgenstern H, Hung
RJ, Christiani DC. Tobacco smoking as a risk factor of bronchioloalveolar
carcinoma of the lung: pooled analysis of seven case-control studies in the
International Lung Cancer Consortium (ILCCO). Cancer Causes Control.
2011;22(1):73-9. PMC3002160.
Chang CM, Wang SS, Dave BJ, Jain S, Vasef MA, Weisenburger DD, Cozen
W, Davis S, Severson RK, Lynch CF, Rothman N, Cerhan JR, Hartge P,
Morton LM. Risk factors for non-Hodgkin lymphoma subtypes defined
by histology and t(14;18) in a population-based case-control study. Int J
Cancer. 2011;129(4):938-47. PMC3125462.
Colt JS, Schwartz K, Graubard BI, Davis F, Ruterbusch J, DiGaetano R,
Purdue M, Rothman N, Wacholder S, Chow WH. Hypertension and risk
of renal cell carcinoma among white and black Americans. Epidemiology.
2011;22(6):797-804. PMC3188386.
Cote ML, Haddad R, Edwards DJ, Atikukke G, Gadgeel S, Soubani AO,
Lonardo F, Bepler G, Schwartz AG, Ethier SP. Frequency and type of
epidermal growth factor receptor mutations in African-Americans
with non-small cell lung cancer. J Thorac Oncol. 2011;6(3):627-30.
PMC3057407.
Daniel CR, Schwartz KL, Colt JS, Dong LM, Ruterbusch JJ, Purdue MP,
Cross AJ, Rothman N, Davis FG, Wacholder S, Graubard BI, Chow WH,
Sinha R. Meat-cooking mutagens and risk of renal cell carcinoma. Br J
Cancer. 2011;105(7):1096-104. PMC3185955.
Filson CP, Miller DC, Colt JS, Ruterbusch J, Linehan WM, Chow WH,
Schwartz K. Surgical approach and the use of lymphadenectomy and
adrenalectomy among patients undergoing radical nephrectomy for renal
cell carcinoma. Urol Oncol. 2011. PMC3123686. [Epub ahead of print]
Gold LS, Stewart PA, Milliken K, Purdue M, Severson R, Seixas N, Blair
A, Hartge P, Davis S, De Roos AJ. The relationship between multiple
myeloma and occupational exposure to six chlorinated solvents. Occup
Environ Med. 2011;68(6):391-9. PMC3094509.
Harlan LC, Lynch CF, Keegan TH, Hamilton AS, Wu XC, Kato I, West MM,
Cress RD, Schwartz SM, Smith AW, Deapen D, Stringer SM, Potosky AL,
Group AHSC. Recruitment and follow-up of adolescent and young adult
29
Metropolitan Detroit Cancer
Surveillance System/SEER Publications 2010-2012 (continued)
cancer survivors: the AYA HOPE Study. J Cancer Surviv. 2011;5(3):30514. PMC3159756.
Hinch AG, Tandon A, Patterson N, Song Y, Rohland N, Palmer CD, Chen GK,
Wang K, Buxbaum SG, Akylbekova EL, Aldrich MC, Ambrosone CB, Amos
C, Bandera EV, Berndt SI, Bernstein L, Blot WJ, Bock CH, Boerwinkle E,
Cai Q, Caporaso N, Casey G, Cupples LA, Deming SL, Diver WR, Divers J,
Fornage M, Gillanders EM, Glessner J, Harris CC, Hu JJ, Ingles SA, Isaacs
W, John EM, Kao WH, Keating B, Kittles RA, Kolonel LN, Larkin E, Le
Marchand L, McNeill LH, Millikan RC, Murphy A, Musani S, NeslundDudas C, Nyante S, Papanicolaou GJ, Press MF, Psaty BM, Reiner AP, Rich
SS, Rodriguez-Gil JL, Rotter JI, Rybicki BA, Schwartz AG, Signorello LB,
Spitz M, Strom SS, Thun MJ, Tucker MA, Wang Z, Wiencke JK, Witte
JS, Wrensch M, Wu X, Yamamura Y, Zanetti KA, Zheng W, Ziegler RG,
Zhu X, Redline S, Hirschhorn JN, Henderson BE, Taylor HA, Jr., Price
AL, Hakonarson H, Chanock SJ, Haiman CA, Wilson JG, Reich D, Myers
SR. The landscape of recombination in African-Americans. Nature.
2011;476(7359):170-5. PMC3154982.
30
Hofmann JN, Baccarelli A, Schwartz K, Davis FG, Ruterbusch JJ, Hoxha M,
McCarthy BJ, Savage SA, Wacholder S, Rothman N, Graubard BI, Colt JS,
Chow WH, Purdue MP. Risk of renal cell carcinoma in relation to blood
telomere length in a population-based case-control study. Br J Cancer.
2011;105(11):1772-5. PMC3242602.
Lan Q, Wang SS, Menashe I, Armstrong B, Zhang Y, Hartge P, Purdue
MP, Holford TR, Morton LM, Kricker A, Cerhan JR, Grulich A, Cozen
W, Zahm SH, Yeager M, Vajdic CM, Schenk M, Leaderer B, Yuenger J,
Severson RK, Chatterjee N, Chanock SJ, Zheng T, Rothman N. Genetic
variation in Th1/Th2 pathway genes and risk of non-Hodgkin lymphoma:
a pooled analysis of three population-based case-control studies. Br J
Haematol. 2011;153(3):341-50. PMC3075370.
Lu Y, Abdou AM, Cerhan JR, Morton LM, Severson RK, Davis S, Cozen
W, Rothman N, Bernstein L, Chanock S, Hartge P, Wang SS. Human
leukocyte antigen class I and II alleles and overall survival in diffuse
large B-cell lymphoma and follicular lymphoma. ScientificWorldJournal.
2011;11:2062-70. PMC3217596.
Mujahid MS, Janz NK, Hawley ST, Griggs JJ, Hamilton AS, Graff J, Katz
SJ. Racial/ethnic differences in job loss for women with breast cancer. J
Cancer Surviv. 2011;5(1):102-11. PMC3040347.
Neklason DW, Done MW, Sargent NR, Schwartz AG, Anton-Culver H,
Griffin CA, Ahnen DJ, Schildkraut JM, Tomlinson GE, Strong LC, Miller
AR, Stopfer JE, Burt RW. Activating mutation in MET oncogene in
familial colorectal cancer. BMC Cancer. 2011;11:424. PMC3202244.
Peterson L, Soliman A, Ruterbusch JJ, Smith N, Schwartz K. Comparison
of exposures among Arab American and non-Hispanic White female
thyroid cancer cases in metropolitan Detroit. J Immigr Minor Health.
2011;13(6):1033-40.
Purdue MP, Bakke B, Stewart P, De Roos AJ, Schenk M, Lynch CF, Bernstein
L, Morton LM, Cerhan JR, Severson RK, Cozen W, Davis S, Rothman
N, Hartge P, Colt JS. A case-control study of occupational exposure to
trichloroethylene and non-Hodgkin lymphoma. Environ Health Perspect.
2011;119(2):232-8.
Purdue MP, Johansson M, Zelenika D, Toro JR, Scelo G, Moore LE,
Prokhortchouk E, Wu X, Kiemeney LA, Gaborieau V, Jacobs KB, Chow
WH, Zaridze D, Matveev V, Lubinski J, Trubicka J, Szeszenia-Dabrowska
N, Lissowska J, Rudnai P, Fabianova E, Bucur A, Bencko V, Foretova L,
Janout V, Boffetta P, Colt JS, Davis FG, Schwartz KL, Banks RE, Selby
PJ, Harnden P, Berg CD, Hsing AW, Grubb RL, 3rd, Boeing H, Vineis P,
Clavel-Chapelon F, Palli D, Tumino R, Krogh V, Panico S, Duell EJ, Quiros
JR, Sanchez MJ, Navarro C, Ardanaz E, Dorronsoro M, Khaw KT, Allen
NE, Bueno-de-Mesquita HB, Peeters PH, Trichopoulos D, Linseisen J,
Ljungberg B, Overvad K, Tjonneland A, Romieu I, Riboli E, Mukeria
A, Shangina O, Stevens VL, Thun MJ, Diver WR, Gapstur SM, Pharoah
PD, Easton DF, Albanes D, Weinstein SJ, Virtamo J, Vatten L, Hveem
K, Njolstad I, Tell GS, Stoltenberg C, Kumar R, Koppova K, Cussenot O,
Benhamou S, Oosterwijk E, Vermeulen SH, Aben KK, van der Marel SL,
Ye Y, Wood CG, Pu X, Mazur AM, Boulygina ES, Chekanov NN, Foglio
M, Lechner D, Gut I, Heath S, Blanche H, Hutchinson A, Thomas G,
Wang Z, Yeager M, Fraumeni JF, Jr., Skryabin KG, McKay JD, Rothman
N, Chanock SJ, Lathrop M, Brennan P. Genome-wide association study
of renal cell carcinoma identifies two susceptibility loci on 2p21 and
11q13.3. Nat Genet. 2011;43(1):60-5. PMC3049257.
Purdue MP, Colt JS, Graubard B, Davis F, Ruterbusch JJ, Digaetano R,
Karami S, Wacholder S, Schwartz K, Chow WH. A case-control study of
reproductive factors and renal cell carcinoma among black and white
women in the United States. Cancer Causes Control. 2011;22(11):153744.
Reinersman JM, Johnson ML, Riely GJ, Chitale DA, Nicastri AD, Soff
GA, Schwartz AG, Sima CS, Ayalew G, Lau C, Zakowski MF, Rusch VW,
Ladanyi M, Kris MG. Frequency of EGFR and KRAS mutations in lung
adenocarcinomas in African-Americans. J Thorac Oncol. 2011;6(1):28-31.
Schwartz AG, Wenzlaff AS, Bock CH, Ruterbusch JJ, Chen W, Cote ML,
Artis AS, Van Dyke AL, Land SJ, Harris CC, Pine SR, Spitz MR, Amos CI,
Levin AM, McKeigue PM. Admixture mapping of lung cancer in 1812
African-Americans. Carcinogenesis. 2011;32(3):312-7. PMC3047238.
Smedby KE, Foo JN, Skibola CF, Darabi H, Conde L, Hjalgrim H, Kumar V,
Chang ET, Rothman N, Cerhan JR, Brooks-Wilson AR, Rehnberg E, Irwan
ID, Ryder LP, Brown PN, Bracci PM, Agana L, Riby J, Cozen W, Davis S,
Hartge P, Morton LM, Severson RK, Wang SS, Slager SL, Fredericksen
ZS, Novak AJ, Kay NE, Habermann TM, Armstrong B, Kricker A, Milliken
S, Purdue MP, Vajdic CM, Boyle P, Lan Q, Zahm SH, Zhang Y, Zheng T,
Leach S, Spinelli JJ, Smith MT, Chanock SJ, Padyukov L, Alfredsson L,
Klareskog L, Glimelius B, Melbye M, Liu ET, Adami HO, Humphreys
K, Liu J. GWAS of follicular lymphoma reveals allelic heterogeneity at
6p21.32 and suggests shared genetic susceptibility with diffuse large
B-cell lymphoma. PLoS Genet. 2011;7(4):e1001378. PMC3080853.
Wang SS, Menashe I, Cerhan JR, Cozen W, Severson RK, Davis S,
Hutchinson A, Rothman N, Chanock SJ, Bernstein L, Hartge P, Morton
LM. Variations in chromosomes 9 and 6p21.3 with risk of non-Hodgkin
lymphoma. Cancer Epidemiol Biomarkers Prev. 2011;20(1):42-9.
Wang SS, Lu Y, Rothman N, Abdou AM, Cerhan JR, De Roos A, Davis S,
Severson RK, Cozen W, Chanock SJ, Bernstein L, Morton LM, Hartge
P. Variation in effects of non-Hodgkin lymphoma risk factors according
to the human leukocyte antigen (HLA)-DRB1*01:01 allele and ancestral
haplotype 8.1. PLoS One. 2011;6(11):e26949. PMC3212525.
Wheeler DC, De Roos AJ, Cerhan JR, Morton LM, Severson R, Cozen W,
Ward MH. Spatial-temporal analysis of non-Hodgkin lymphoma in
the NCI-SEER NHL case-control study. Environ Health. 2011;10:63.
PMC3148953.
2010
Amos CI, Pinney SM, Li Y, Kupert E, Lee J, de Andrade MA, Yang P,
Schwartz AG, Fain PR, Gazdar A, Minna J, Wiest JS, Zeng D, Rothschild
H, Mandal D, You M, Coons T, Gaba C, Bailey-Wilson JE, Anderson MW.
A susceptibility locus on chromosome 6q greatly increases lung cancer
risk among light and never smokers. Cancer Res. 2010;70(6):2359-67.
PMC2855643.
Boleij A, Roelofs R, Schaeps RM, Schulin T, Glaser P, Swinkels DW, Kato
I, Tjalsma H. Increased exposure to bacterial antigen RpL7/L12 in
early stage colorectal cancer patients. Cancer. 2010;116(17):4014-22.
PMC2930125.
Cetin K, Beebe-Dimmer JL, Fryzek JP, Markus R, Carducci MA. Recent
time trends in the epidemiology of stage IV prostate cancer in the United
States: analysis of data from the Surveillance, Epidemiology, and End
Results Program. Urology. 2010;75(6):1396-404.
De Roos AJ, Davis S, Colt JS, Blair A, Airola M, Severson RK, Cozen W,
Cerhan JR, Hartge P, Nuckols JR, Ward MH. Residential proximity to
industrial facilities and risk of non-Hodgkin lymphoma. Environ Res.
2010;110(1):70-8. PMC2795078.
Dombi GW, Rosbolt JP, Severson RK. Neural network analysis of
employment history as a risk factor for prostate cancer. Comput Biol
Med. 2010;40(9):751-7.
Fang S, Pinney SM, Bailey-Wilson JE, de Andrade MA, Li Y, Kupert E, You
M, Schwartz AG, Yang P, Anderson MW, Amos CI. Ordered subset analysis
identifies loci influencing lung cancer risk on chromosomes 6q and 12q.
Cancer Epidemiol Biomarkers Prev. 2010;19(12):3157-66.
Geyer SM, Morton LM, Habermann TM, Allmer C, Davis S, Cozen W,
Severson RK, Lynch CF, Wang SS, Maurer MJ, Hartge P, Cerhan JR.
Smoking, alcohol use, obesity, and overall survival from non-Hodgkin
lymphoma: a population-based study. Cancer. 2010;116(12):2993-3000.
PMC2889918.
Gold LS, Milliken K, Stewart P, Purdue M, Severson R, Seixas N, Blair A,
Davis S, Hartge P, De Roos AJ. Occupation and multiple myeloma: an
occupation and industry analysis. Am J Ind Med. 2010;53(8):768-79.
Grulich AE, Vajdic CM, Falster MO, Kane E, Smedby KE, Bracci PM, de
Sanjose S, Becker N, Turner J, Martinez-Maza O, Melbye M, Engels EA,
Vineis P, Costantini AS, Holly EA, Spinelli JJ, La Vecchia C, Zheng T, Chiu
BC, Franceschi S, Cocco P, Maynadie M, Foretova L, Staines A, Brennan P,
Davis S, Severson RK, Cerhan JR, Breen EC, Birmann B, Cozen W. Birth
order and risk of non-hodgkin lymphoma--true association or bias? Am J
Epidemiol. 2010;172(6):621-30. PMC2950815.
Jagsi R, Abrahamse P, Morrow M, Hawley ST, Griggs JJ, Graff JJ, Hamilton
AS, Katz SJ. Patterns and correlates of adjuvant radiotherapy receipt
after lumpectomy and after mastectomy for breast cancer. J Clin Oncol.
2010;28(14):2396-403.
Kakarala M, Rozek L, Cote M, Liyanage S, Brenner DE. Breast cancer
histology and receptor status characterization in Asian Indian and
Pakistani women in the U.S.--a SEER analysis. BMC Cancer. 2010;10:191.
PMC2873947.
Karami S, Schwartz K, Purdue MP, Davis FG, Ruterbusch JJ, Munuo SS,
Wacholder S, Graubard BI, Colt JS, Chow WH. Family history of cancer
and renal cell cancer risk in Whites and African-Americans. Br J Cancer.
2010;102(11):1676-80. PMC2883149.
Kato I, Land S, Majumdar AP, Barnholtz-Sloan J, Severson RK. Functional
polymorphisms to modulate luminal lipid exposure and risk of colorectal
cancer. Cancer Epidemiol. 2010;34(3):291-7. PMC2905870.
Kato I, Majumdar AP, Land SJ, Barnholtz-Sloan JS, Severson RK. Dietary
fatty acids, luminal modifiers, and risk of colorectal cancer. Int J Cancer.
2010;127(4):942-51. PMC2891322.
Katz SJ, Hawley ST, Morrow M, Griggs JJ, Jagsi R, Hamilton AS, Graff
JJ, Friese CR, Hofer TP. Coordinating cancer care: patient and practice
management processes among surgeons who treat breast cancer. Med
Care. 2010;48(1):45-51.
Katz SJ, Hawley ST, Abrahamse P, Morrow M, Friese CR, Alderman AK,
Griggs JJ, Hamilton AS, Graff JJ, Hofer TP. Does it matter where you go
for breast surgery?: attending surgeon’s influence on variation in receipt
of mastectomy for breast cancer. Med Care. 2010;48(10):892-9.
Liu P, Yang P, Wu X, Vikis HG, Lu Y, Wang Y, Schwartz AG, Pinney SM, de
Andrade M, Gazdar A, Gaba C, Mandal D, Lee J, Kupert E, Seminara D,
Minna J, Bailey-Wilson JE, Spitz M, Amos CI, Anderson MW, You M. A
second genetic variant on chromosome 15q24-25.1 associates with lung
cancer. Cancer Res. 2010;70(8):3128-35.
Liu P, Vikis HG, Lu Y, Wang Y, Schwartz AG, Pinney SM, Yang P, de Andrade
M, Gazdar A, Gaba C, Mandal D, Lee J, Kupert E, Seminara D, Minna J,
Bailey-Wilson JE, Amos CI, Anderson MW, You M. Cumulative effect
of multiple loci on genetic susceptibility to familial lung cancer. Cancer
Epidemiol Biomarkers Prev. 2010;19(2):517-24. PMC2846747.
McWalter K, Graham L, Atzinger C. Presented Abstracts from the TwentyNinth Annual Education Conference of the National Society of Genetic
Counselors (Dallas, Texas, October 2010) (Abstract by Kennelly, K.,
Salkowski, A., Schwartz, AG.: Preliminary Response to the Re-consent
Process for Inclusion in dbGaP). Journal of Genetic Counseling.
2010;19(6):653.
Miller DC, Ruterbusch J, Colt JS, Davis FG, Linehan WM, Chow
WH, Schwartz K. Contemporary clinical epidemiology of renal cell
carcinoma: insight from a population based case-control study. J Urol.
2010;184(6):2254-8.
Nahleh Z, Abrams J, Bhargaval A, Nirmal K, Graff JJ. Outcome of patients
with early breast cancer receiving nitrogen-containing bisphosphonates:
a comparative analysis from the metropolitan detroit cancer surveillance
system. Clin Breast Cancer. 2010;10(6):459-64.
Shen M, Menashe I, Morton LM, Zhang Y, Armstrong B, Wang SS, Lan Q,
Hartge P, Purdue MP, Cerhan JR, Grulich A, Cozen W, Yeager M, Holford
TR, Vajdic CM, Davis S, Leaderer B, Kricker A, Severson RK, Zahm SH,
Chatterjee N, Rothman N, Chanock SJ, Zheng T. Polymorphisms in DNA
repair genes and risk of non-Hodgkin lymphoma in a pooled analysis of
three studies. Br J Haematol. 2010;151(3):239-44. PMC2967772.
Truong T, Hung RJ, Amos CI, Wu X, Bickeboller H, Rosenberger A, Sauter
W, Illig T, Wichmann HE, Risch A, Dienemann H, Kaaks R, Yang P, Jiang
R, Wiencke JK, Wrensch M, Hansen H, Kelsey KT, Matsuo K, Tajima K,
Schwartz AG, Wenzlaff A, Seow A, Ying C, Staratschek-Jox A, Nurnberg
P, Stoelben E, Wolf J, Lazarus P, Muscat JE, Gallagher CJ, Zienolddiny
S, Haugen A, van der Heijden HF, Kiemeney LA, Isla D, Mayordomo
JI, Rafnar T, Stefansson K, Zhang ZF, Chang SC, Kim JH, Hong YC,
Duell EJ, Andrew AS, Lejbkowicz F, Rennert G, Muller H, Brenner H, Le
Marchand L, Benhamou S, Bouchardy C, Teare MD, Xue X, McLaughlin
J, Liu G, McKay JD, Brennan P, Spitz MR. Replication of lung cancer
susceptibility loci at chromosomes 15q25, 5p15, and 6p21: a pooled
analysis from the International Lung Cancer Consortium. J Natl Cancer
Inst. 2010;102(13):959-71. PMC2897877.
Wang SS, Abdou AM, Morton LM, Thomas R, Cerhan JR, Gao X, Cozen W,
Rothman N, Davis S, Severson RK, Bernstein L, Hartge P, Carrington M.
Human leukocyte antigen class I and II alleles in non-Hodgkin lymphoma
etiology. Blood. 2010;115(23):4820-3. PMC2890176.
31
Affiliations and Acknowledgements
STATE OF MICHIGAN
The cancer statistics collected and disseminated here could not have
been presented without the untiring work of the cancer registrar,
editing, follow-up and management teams of the Metropolitan
Detroit Cancer Surveillance System. Nor could this work have been
presented without the collaboration of each of the facilities on the
following page and the entities listed here.
32
Special thanks go to the Michigan Department of Community
Health for their ongoing assistance, and also to the many physicians
who support our efforts to ensure complete cancer reporting for
tri-county residents. Finally, we would like to acknowledge the
cancer patients and their families. It is to you we dedicate this work.
The Metropolitan Detroit Cancer Surveillance System is funded by:
The Division of Cancer Prevention and Control
National Cancer Institute
Department of Health and Human Services
SEER Contract # HHSN261201000028C
Control # NO1-PC-00028
Metropolitan Detroit Cancer Surveillance System
Participating Institutions
The following hospitals, pathology laboratories and radiation
oncology treatment facilities are participants in the Metropolitan
Detroit Cancer Surveillance System. Their collaboration makes
possible Detroit area population-based cancer reporting.
Hospitals and Associated Path Labs, Radiation Facilities and Clinics
Botsford General Hospital
Hutzel Hospital
Providence Hospital
Children’s Hospital of Michigan
John D. Dingell Dept. of Veterans
Administration Medical Center
St. John Detroit Riverview Hospital
Crittenton Hospital
Detroit Receiving Hospital
Doctors Hospital of Michigan
Garden City Hospital
Harper Hospital
University Health Center
Karmanos Cancer Center
St. John Hospital and Medical
Center
McLaren Hospital – Macomb
St. John Macomb Hospital Center
McLaren Hospital – Oakland
St. John Northshore Hospital
Michigan Orthopedic Specialty
Hospital
St. John Oakland Hospital
William Beaumont Hospital –
Grosse Pointe
William Beaumont Hospital –
Royal Oak
William Beaumont Hospital –
Troy
Henry Ford Hospital
Oakwood Hospital
St. Joseph Mercy Hospital
Ann Arbor*
Henry Ford – Warren
Oakwood Annapolis Hospital
St. Joseph Mercy Hospital Oakland
Henry Ford – Macomb
Oakwood Heritage Hospital
St. Mary Hospital
Henry Ford Wyandotte Hospital
Oakwood Southshore Medical
Center
Sinai-Grace Hospital
Huron Valley-Sinai Hospital
Southeast Michigan Surgical
Hospital
University Hospital Ann Arbor*
Veterans Administration Medical
Center – Ann Arbor*
Independent Pathology Laboratories
Ameripath Institute of Urology
Detroit Bio-Medical Laboratories
Medical Diagnostic Lab
Pinkus Laboratory
Beaumont Reference Laboratory
Dianon Systems
Michigan Institute of Urology
Plus Diagnostics
Bio-Reference Laboratory
Genoptix Laboratory
Midwest Skin Pathology
Quest Diagnostics, Inc.
Bostwick Laboratories
Hilbrich Dermatology Lab, PLC
Novice Dermatopathology Lab
St. John Oral Pathology
Contemporary Imaging Associates
Hospital Consolidated Laboratories
Oakwood Medical Laboratory
Dermatopathology Associates
Laboratory Corporation of America
Oppenheimer Urologic Reference
Laboratory
Independent Radiation Oncology Facilities
21st Century Oncology
Downriver Center for Oncology
Radiation Therapy Associates
*Hospital not located in the tri-county area.
33
Appendix A
Michigan Public Health Code and HIPAA (Excerpts)
Michigan Act 368 of 1978 and HIPAA Law and Federal Rules (Excerpts);
Public Law 104-191; 104th Congress; August 21, 1996
333.2619 Cancer registry; establishment; purpose; reports; records;
rules; medical or department examination or supervision not
required; contracts; evaluation of reports; publication of summary
reports; commencement of reporting; effective date of section.
and make available to the public reports summarizing the
information collected. The first summary report shall be
published not later than 180 days after the end of the first
2 full calendar years after the effective date of this section.
Subsequent annual summary reports shall be made on a full
calendar year basis and published not later than 180 days after
the end of each calendar year.
Sec. 2619.
(1) The department shall establish a registry to record cases
of cancer and other specified tumorous and precancerous
diseases that occur in the state, and to record
information concerning these cases as the department
considers necessary and appropriate in order to conduct
epidemiologic surveys of cancer and cancer-related
diseases in the state.
34
(2) Each diagnosed case of cancer and other specified
tumorous and precancerous diseases shall be reported to
the department pursuant to subsection (4), or reported
to a cancer reporting registry if the cancer reporting
registry meets standards established pursuant to
subsection (4) to ensure the accuracy and completeness of
the reported information. A person or facility required to
report a diagnosis pursuant to subsection (4) may elect to
report the diagnosis to the state through an existing cancer
registry only if the registry meets minimum reporting
standards established by the department.
(3) The department shall maintain comprehensive records
of all reports submitted pursuant to this section. These reports
shall be subject to the same requirements of confidentiality as
provided in section 2631 for data or records concerning medical
research projects.
(4) The director shall promulgate rules which provide for
all of the following:
(8) Reporting pursuant to subsection (2) shall begin the next
calendar year after the effective date of this section.
(9) This section shall take effect July 1, 1984.
History: Add. 1984, Act 82, Eff. July 1, 1984
Popular Name: Act 368
333.2621 Comprehensive policy for conduct and support of
research and demonstration activities; conducting and supporting
demonstration projects and scientific evaluations.
Sec. 2621.
(1) The department shall establish a comprehensive policy
pursuant to and consistent with section 2611(2) for the
conduct and support of research and demonstration activities
related to the department’s responsibility for the health care
needs of the people of this state.
(2) The department shall conduct research and demonstration
activities related to the department’s responsibility for the
environmental, preventive, and personal health needs of the
communities and people of this state, including:
(a)The causes, effects, and methods of prevention of illness.
(a) A list of tumorous and precancerous diseases other than
cancer to be reported pursuant to subsection (2).
(b)The determinants of health, including behavior related
to health.
(b) The quality and manner in which the cases and other
information described in subsection (1) are reported
to the department.
(c)The accessibility, acceptability, availability, organization,
distribution, utilization, quality, and financing of health
care, especially those services for the medically needy.
(c) Th
e terms and conditions under which records disclosing the
name and medical condition of a specific individual and kept
pursuant to this section are released by the department.
(3) The department may conduct and support demonstration
projects to carry out subsection (2).
(5) This section does not compel an individual to submit
to medical or department examination or supervision.
(6) The department may contract for the collection and
analysis of, and research related to, the epidemiologic
data required under this section.
(7) Within 2 years after the effective date of this section, the
department shall begin evaluating the reports collected
pursuant to subsection (2). The department shall publish
(4) The department shall conduct or support the conduct of
scientific evaluations of the effectiveness, efficiency, and
relevance of programs conducted or supported by the
department.
History: 1978, Act 368, Eff. Sept. 30, 1978
Popular Name: Act 368
333.2631 Data concerning medical research project;
confidentiality; use.
333.2633 Data concerning medical research projects; liability for
furnishing.
Sec. 2631.
Sec. 2633.
The information, records of interviews, written reports,
statements, notes, memoranda, or other data or records
furnished to, procured by, or voluntarily shared with the
department in the conduct of a medical research project,
or a person, agency, or organization which has been designated
in advance by the department as a medical research project
which regularly furnishes statistical or summary data with
respect to that project to the department for the purpose of
reducing the morbidity or mortality from any cause or condition
of health are confidential and shall be used solely for statistical,
scientific, and medical research purposes relating to the cause
or condition of health.
The furnishing of information, records, reports, statements,
notes, memoranda, or other data to the department, either
voluntarily or as required by this code, or to a person, agency, or
organization designated as a medical research project does not
subject a physician, hospital, sanatorium, rest home, nursing
home, or other person or agency furnishing the information,
records, reports, statements, notes, memoranda, or other data
to liability in an action for damages or other relief, and is not
considered to be the willful betrayal of a professional secret or the
violation of a confidential relationship. History: 1978, Act 368, Eff. Sept. 30, 1978
Popular Name: Act 368
History: 1978, Act 368, Eff. Sept. 30, 1978 ;-- Am. 1988, Act 122, Eff. Mar. 30, 1989
Popular Name: Act 368
35
333.2632 Data concerning medical research project; inadmissible
as evidence; exhibition or disclosure.
Sec. 2632.
The information, records, reports, statements, notes,
memoranda, or other data described in section 2631 are not
admissible as evidence in an action in a court or before any
other tribunal, board, agency, or person. Furnishing the data
to the department in the conduct of a medical research project
or to a designated medical research project does not result in
the loss of any privilege which the data may otherwise have
making them inadmissible as evidence. The information, records,
reports, notes, memoranda, or other data shall not be exhibited
nor their contents disclosed in any way, in whole or in part, by
the department or its representative, or by any other person,
agency, or organization, except as is necessary for the purpose
of furthering the medical research project to which they relate
consistent with section 2637 and the rules promulgated under
section 2678. A person participating in a designated medical
research project shall not disclose the information obtained
except in strict conformity with the research project.
History: 1978, Act 368, Eff. Sept. 30, 1978
Popular Name: Act 368
•••
SEC. 1178. EFFECT ON STATE LAW
(b) PUBLIC HEALTH.--Nothing in this part shall be
construed to invalidate or limit the authority, power, or
procedures established under any law providing for the
reporting of disease or injury, child abuse, birth, or death,
public health surveillance, or public health investigation
or intervention.
(c) STATE REGULATORY REPORTING.--Nothing in this
part shall limit the ability of a State to require a health
plan to report, or to provide access to, information for
management audits, financial audits, program monitoring
and evaluation, facility licensure or certification, or
individual licensure or certification.
•••
Rendered 6/7/2005 16:58:30
Michigan Compiled Laws
© 2005 Legislative Council, State of Michigan
Courtesy of www.legislature.mi.gov
Source: http://aspe.hhs.gov/admnsimp/pl104191.htm (emphasis added)
Metropolitan Detroit Cancer Surveillance System
Epidemiology Section | 4100 John R Street MM04EP | Detroit, MI 48201
For Additional Information:
Telephone: (313) 578-4231
Fax: (313) 578-4306
http://seer.cancer.gov/registries/detroit.html
Prepared By
Contributing Authors
Ron Shore, MPH
Biostatistical Programmer
Michele L. Cote, PhD
Patricia Arballo-Spong, BS
SEER Project Coordinator
Ikuko Kato, MD, PhD, Patricia Arballo-Spong, BS,
Ann S. Bankowski, MSW, Kari A. Borden, RHIT, CTR
Ellen M. Velie, PhD, Kendra Schwartz, MD, MSPH
Fawn D. Vigneau, JD, MPH
Co-Director, Epidemiology Research Core
36
Kendra Schwartz, MD, MSPH
Director, Epidemiology Research Core
Ann G. Schwartz, PhD, MPH
Patricia Arballo-Spong, BS
Ann S. Bankowski, MSW
Ann G. Schwartz, PhD, MPH
Director, Metropolitan Detroit Cancer Surveillance System
Kari A. Borden, RHIT, CTR
Leadership Team
Ann G. Schwartz, PhD, MPH
Director, Metropolitan Detroit Cancer Surveillance System
SEER Principal Investigator
Kendra Schwartz, MD, MSPH
SEER Co-Investigator
Director, Epidemiology Research Core
Fawn D. Vigneau, JD, MPH
Co-Director, Epidemiology Research Core
Patricia Arballo-Spong, BS
SEER Project Coordinator
William O. Quarshie, MS
Biostatistician
Joanne Harris, CTR
Chief of Cancer Surveillance
Nancy L. Lozon, BS, CTR
Manager Cancer Surveillance - Office Operations
Patrick Nicolin, BA, CTR
Manager Cancer Surveillance - Quality Control and Field Operations
Richard Pense, BS
Director, Basic and Population Science Systems
Inhee Han, BS
Senior Applications Analyst
Gloria Kwiatkowski, BS
Senior Applications Analyst
Sharon Moton, BS
Administrator
Published December 2012