1932

Abstract

Racial and ethnic disparities span the continuum of cancer care and are driven by a complex interplay among social, psychosocial, lifestyle, environmental, health system, and biological determinants of health. Research is needed to identify these determinants of cancer health disparities and to develop interventions to achieve cancer health equity. Herein, we focus on the overall burden of ancestry-related molecular alterations, the functional significance of the alterations in hallmarks of cancer, and the implications of the alterations for precision oncology and immuno-oncology. In conclusion, we reflect on the importance of estimating ancestry, improving diverse racial and ethnic participation in cancer clinical trials, and examining the intersection among determinants of cancer health disparities.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-070119-120305
2021-01-27
2024-04-27
Loading full text...

Full text loading...

/deliver/fulltext/med/72/1/annurev-med-070119-120305.html?itemId=/content/journals/10.1146/annurev-med-070119-120305&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Broder S. 1991. Progress and challenges in the National Cancer Program. Origins of Human Cancer: A Comprehensive Review J Brugge, T Curran, E Harlow, F McCormick pp. 27–34 Plainview, NY: Cold Spring Harbor Lab. Press
    [Google Scholar]
  2. 2. 
    Tan DSW, Mok TSK, Rebbeck TR 2016. Cancer genomics: diversity and disparity across ethnicity and geography. J. Clin. Oncol. 34:191–101
    [Google Scholar]
  3. 3. 
    Morton RA Jr 1994. Racial differences in adenocarcinoma of the prostate in North American men. Urology 44:5637–45
    [Google Scholar]
  4. 4. 
    Rose DP, Royak-Schaler R. 2001. Tumor biology and prognosis in black breast cancer patients: a review. Cancer Detect. Prev. 25:116–31
    [Google Scholar]
  5. 5. 
    Lane D. 2016. p53: out of Africa. Genes Dev 30:8876–77
    [Google Scholar]
  6. 6. 
    Shiao Y-H, Chen VW, Scheer WD et al. 1995. Racial disparity in the association of p53 gene alterations with breast cancer survival. Cancer Res 55:71485–90
    [Google Scholar]
  7. 7. 
    Wu ES, Park JY, Zeitouni JA et al. 2016. Effect of actionable somatic mutations on racial/ethnic disparities in head and neck cancer prognosis. Head Neck 38:81234–41
    [Google Scholar]
  8. 8. 
    Zhang W, Edwards A, Flemington EK, Zhang K 2017. Racial disparities in patient survival and tumor mutation burden, and the association between tumor mutation burden and cancer incidence rate. Sci. Rep. 7:113639
    [Google Scholar]
  9. 9. 
    Jaratlerdsiri W, Chan EKF, Gong T et al. 2018. Whole-genome sequencing reveals elevated tumor mutational burden and initiating driver mutations in African men with treatment-naïve, high-risk prostate cancer. Cancer Res 78:246736–46
    [Google Scholar]
  10. 10. 
    Churpek JE, Walsh T, Zheng Y et al. 2015. Inherited predisposition to breast cancer among African American women. Breast Cancer Res. Treat. 149:131–39
    [Google Scholar]
  11. 11. 
    Wright N, Rida P, Krishnamurti U et al. 2017. Targeted drugs and diagnostic assays: companions in the race to combat ethnic disparity. Front. Biosci. 22:193–211
    [Google Scholar]
  12. 12. 
    Anders C, Carey LA. 2008. Understanding and treating triple-negative breast cancer. Oncology 22:111233–39
    [Google Scholar]
  13. 13. 
    Haffty BG, Yang Q, Reiss M et al. 2006. Locoregional relapse and distant metastasis in conservatively managed triple negative early-stage breast cancer. J. Clin. Oncol. 24:365652–57
    [Google Scholar]
  14. 14. 
    Steuer CE, Behera M, Berry L et al. 2016. Role of race in oncogenic driver prevalence and outcomes in lung adenocarcinoma: results from the Lung Cancer Mutation Consortium. Cancer 122:5766–72
    [Google Scholar]
  15. 15. 
    Kwon DH-M, Borno HT, Cheng HH et al. 2019. Ethnic disparities among men with prostate cancer undergoing germline testing. Urol. Oncol. 38:380.e1–80.e7
    [Google Scholar]
  16. 16. 
    Spratt DE, Chan T, Waldron L et al. 2016. Racial/ethnic disparities in genomic sequencing. JAMA Oncol 2:81070–74
    [Google Scholar]
  17. 17. 
    Jia F, Teer JK, Knepper TC et al. 2017. Discordance of somatic mutations between Asian and Caucasian patient populations with gastric cancer. Mol. Diagn. Ther. 21:2179–85
    [Google Scholar]
  18. 18. 
    Aurias A, Rimbaut C, Buffe D et al. 1983. Chromosomal translocations in Ewing's sarcoma. N. Engl. J. Med. 309:8496–98
    [Google Scholar]
  19. 19. 
    Blackburn J, Vecchiarelli S, Heyer EE et al. 2019. TMPRSS2-ERG fusions linked to prostate cancer racial health disparities: a focus on Africa. Prostate 79:101191–96
    [Google Scholar]
  20. 20. 
    Jiang H, Mao X, Huang X et al. 2016. TMPRSS2:ERG fusion gene occurs less frequently in Chinese patients with prostate cancer. Tumour Biol 37:912397–402 Erratum. 2016 Tumour Biol 37:1014331
    [Google Scholar]
  21. 21. 
    Zhou CK, Young D, Yeboah ED et al. 2017. TMPRSS2:ERG gene fusions in prostate cancer of West African men and a meta-analysis of racial differences. Am. J. Epidemiol. 186:121352–61
    [Google Scholar]
  22. 22. 
    Shinmura K, Kahyo T, Kato H et al. 2014. RSPO fusion transcripts in colorectal cancer in Japanese population. Mol. Biol. Rep. 41:85375–84
    [Google Scholar]
  23. 23. 
    Lee TI, Young RA. 2013. Transcriptional regulation and its misregulation in disease. Cell 152:61237–51
    [Google Scholar]
  24. 24. 
    Bradner JE, Hnisz D, Young RA 2017. Transcriptional addiction in cancer. Cell 168:4629–43
    [Google Scholar]
  25. 25. 
    van ’t Veer LJ, Dai H, van de Vijver MJ et al. 2002. Gene expression profiling predicts clinical outcome of breast cancer. Nature 415:6871530–36
    [Google Scholar]
  26. 26. 
    Grunda JM, Steg AD, He Q et al. 2012. Differential expression of breast cancer-associated genes between stage- and age-matched tumor specimens from African- and Caucasian-American women diagnosed with breast cancer. BMC Res. Notes 5:248
    [Google Scholar]
  27. 27. 
    Allard JE, Chandramouli GVR, Stagliano K et al. 2012. Analysis of PSPHL as a candidate gene influencing the racial disparity in endometrial cancer. Front. Oncol. 2:65
    [Google Scholar]
  28. 28. 
    Jovov B, Araujo-Perez F, Sigel CS et al. 2012. Differential gene expression between African American and European American colorectal cancer patients. PLOS ONE 7:1e30168
    [Google Scholar]
  29. 29. 
    Mitchell KA, Zingone A, Toulabi L et al. 2017. Comparative transcriptome profiling reveals coding and noncoding RNA differences in NSCLC from African Americans and European Americans. Clin. Cancer Res. 23:237412–25
    [Google Scholar]
  30. 30. 
    Powell IJ, Dyson G, Land S et al. 2013. Genes associated with prostate cancer are differentially expressed in African American and European American men. Cancer Epidemiol. Biomarkers Prev. 22:5891–97
    [Google Scholar]
  31. 31. 
    D'Arcy M, Fleming J, Robinson WR et al. 2015. Race-associated biological differences among Luminal A breast tumors. Breast Cancer Res. Treat. 152:2437–48
    [Google Scholar]
  32. 32. 
    Barrow MA, Martin ME, Coffey A et al. 2019. A functional role for the cancer disparity-linked genes, CRYβB2 and CRYβB2P1, in the promotion of breast cancer. Breast Cancer Res 21:1105
    [Google Scholar]
  33. 33. 
    Jiang YZ, Ma D, Suo C et al. 2019. Genomic and transcriptomic landscape of triple-negative breast cancers: subtypes and treatment strategies. Cancer Cell 35:3428–40.e5
    [Google Scholar]
  34. 34. 
    Heiser LM, Mills GB, Gray JW 2019. Therapeutic clues from an integrated omic assessment of East Asian triple negative breast cancers. Cancer Cell 35:3341–43
    [Google Scholar]
  35. 35. 
    Wang ET, Sandberg R, Luo S et al. 2008. Alternative isoform regulation in human tissue transcriptomes. Nature 456:7221470–76
    [Google Scholar]
  36. 36. 
    Pandya-Jones A. 2011. Pre-mRNA splicing during transcription in the mammalian system. Wiley Interdiscip. Rev. RNA 2:5700–17
    [Google Scholar]
  37. 37. 
    Black DL. 2003. Mechanisms of alternative pre-messenger RNA splicing. Annu. Rev. Biochem. 72:291–336
    [Google Scholar]
  38. 38. 
    Graveley BR. 2001. Alternative splicing: increasing diversity in the proteomic world. Trends Genet 17:2100–7
    [Google Scholar]
  39. 39. 
    Baralle FE, Giudice J. 2017. Alternative splicing as a regulator of development and tissue identity. Nat. Rev. Mol. Cell Biol. 18:7437–51
    [Google Scholar]
  40. 40. 
    Pal S, Gupta R, Davuluri RV 2012. Alternative transcription and alternative splicing in cancer. Pharmacol. Ther. 136:3283–94
    [Google Scholar]
  41. 41. 
    Singh B, Eyras E. 2017. The role of alternative splicing in cancer. Transcription 8:291–98
    [Google Scholar]
  42. 42. 
    El Marabti E, Younis I 2018. The cancer spliceome: reprograming of alternative splicing in cancer. Front. Mol. Biosci. 5:80
    [Google Scholar]
  43. 43. 
    Wang BD, Ceniccola K, Hwang S et al. 2017. Alternative splicing promotes tumour aggressiveness and drug resistance in African American prostate cancer. Nat. Commun 8:15921 Corrigendum. 2017 Nat. Commun 27:816161
    [Google Scholar]
  44. 44. 
    Frampton GM, Ali SM, Rosenzweig M et al. 2015. Activation of MET via diverse exon 14 splicing alterations occurs in multiple tumor types and confers clinical sensitivity to MET inhibitors. Cancer Discov 5:8850–59
    [Google Scholar]
  45. 45. 
    Reungwetwattana T, Liang Y, Zhu V, Ou SI 2017. The race to target MET exon 14 skipping alterations in non-small cell lung cancer: the why, the how, the who, the unknown, and the inevitable. Lung Cancer 103:27–37
    [Google Scholar]
  46. 46. 
    Teslow EA, Bao B, Dyson G et al. 2018. Exogenous IL-6 induces mRNA splice variant MBD2_v2 to promote stemness in TP53 wild-type, African American PCa cells. Mol. Oncol. 12:71138–52
    [Google Scholar]
  47. 47. 
    Teslow EA, Mitrea C, Bao B et al. 2019. Obesity-induced MBD2_v2 expression promotes tumor-initiating triple-negative breast cancer stem cells. Mol. Oncol. 13:4894–908
    [Google Scholar]
  48. 48. 
    Calin GA, Croce CM. 2006. MicroRNA signatures in human cancers. Nat. Rev. Cancer 6:11857–66
    [Google Scholar]
  49. 49. 
    Jiang S, Cheng SJ, Ren LC et al. 2019. An expanded landscape of human long noncoding RNA. Nucleic Acids Res 47:157842–56
    [Google Scholar]
  50. 50. 
    Bovell LC, Shanmugam C, Putcha BD et al. 2013. The prognostic value of microRNAs varies with patient race/ethnicity and stage of colorectal cancer. Clin. Cancer Res. 19:143955–65
    [Google Scholar]
  51. 51. 
    Jones J, Grizzle W, Wang H, Yates C 2013. MicroRNAs that affect prostate cancer: emphasis on prostate cancer in African Americans. Biotech. Histochem. 88:7410–24
    [Google Scholar]
  52. 52. 
    Wang BD, Ceniccola K, Yang Q et al. 2015. Identification and functional validation of reciprocal microRNA-mRNA pairings in African American prostate cancer disparities. Clin. Cancer Res. 21:214970–84
    [Google Scholar]
  53. 53. 
    Shao J, Fang PH, He B et al. 2016. Downregulated microRNA-133a in gastric juice as a clinicopathological biomarker for gastric cancer screening. Asian Pac. J. Cancer Prev. 17:52719–22
    [Google Scholar]
  54. 54. 
    Zhang S, Zhau HE, Osunkoya AO et al. 2010. Vascular endothelial growth factor regulates myeloid cell leukemia-1 expression through neuropilin-1-dependent activation of c-MET signaling in human prostate cancer cells. Mol. Cancer 9:9
    [Google Scholar]
  55. 55. 
    Liu C, Chen L, You Z et al. 2020. Association between lncRNA H19 polymorphisms and cancer susceptibility based on a meta-analysis from 25 studies. Gene 729:144317
    [Google Scholar]
  56. 56. 
    Raveh E, Matouk IJ, Gilon M, Hochberg A 2015. The H19 long non-coding RNA in cancer initiation, progression and metastasis—a proposed unifying theory. Mol. Cancer 14:184
    [Google Scholar]
  57. 57. 
    Ahmad A, Azim S, Zubair H et al. 2017. Epigenetic basis of cancer health disparities: looking beyond genetic differences. Biochim. Biophys. Acta Rev. Cancer 1868:116–28
    [Google Scholar]
  58. 58. 
    Chen E, Miller GE, Yu T, Brody GH 2016. The Great Recession and health risks in African American youth. Brain Behav. Immun. 53:234–241
    [Google Scholar]
  59. 59. 
    Miller GE, Yu T, Chen E, Brody GH 2015. Self-control forecasts better psychosocial outcomes but faster epigenetic aging in low-SES youth. PNAS 112:3310325–30
    [Google Scholar]
  60. 60. 
    Janusek LW, Tell D, Gaylord-Harden N, Mathews HL 2017. Relationship of childhood adversity and neighborhood violence to a proinflammatory phenotype in emerging adult African American men: an epigenetic link. Brain Behav. Immun. 60:126–35
    [Google Scholar]
  61. 61. 
    Mehrotra J, Ganpat MM, Kanaan Y et al. 2004. Estrogen receptor/progesterone receptor-negative breast cancers of young African-American women have a higher frequency of methylation of multiple genes than those of Caucasian women. Clin. Cancer Res. 10:62052–57
    [Google Scholar]
  62. 62. 
    Nelson WG, De Marzo AM, Isaacs WB 2003. Prostate cancer. N. Engl. J. Med. 349:4366–81
    [Google Scholar]
  63. 63. 
    Kwabi-Addo B, Wang S, Chung W et al. 2010. Identification of differentially methylated genes in normal prostate tissues from African American and Caucasian men. Clin. Cancer Res. 16:143539–47
    [Google Scholar]
  64. 64. 
    Palucka AK, Coussens LM. 2016. The basis of oncoimmunology. Cell 164:61233–47
    [Google Scholar]
  65. 65. 
    Sun G, Dong X, Tang X et al. 2019. The prognostic value of immunoscore in patients with colorectal cancer: a systematic review and meta-analysis. Cancer Med 8:1182–89
    [Google Scholar]
  66. 66. 
    Paulucci DJ, Sfakianos JP, Skanderup AJ et al. 2017. Genomic differences between black and white patients implicate a distinct immune response to papillary renal cell carcinoma. Oncotarget 8:35196–205
    [Google Scholar]
  67. 67. 
    Jenkins BD, Martini RN, Hire R et al. 2019. Atypical chemokine receptor 1 (DARC/ACKR1) in breast tumors is associated with survival, circulating chemokines, tumor-infiltrating immune cells, and African ancestry. Cancer Epidemiol. Biomarkers Prev. 28:4690–700
    [Google Scholar]
  68. 68. 
    Yeyeodu ST, Kidd LR, Kimbro KS 2019. Protective innate immune variants in racial/ethnic disparities of breast and prostate cancer. Cancer Immunol. Res. 7:91384–89
    [Google Scholar]
  69. 69. 
    Kaur HB, Lu J, Guedes LB et al. 2019. TP53 missense mutation is associated with increased tumor-infiltrating T cells in primary prostate cancer. Hum. Pathol. 87:95–102
    [Google Scholar]
  70. 70. 
    Pritchard AL. 2018. Targeting neoantigens for personalised immunotherapy. BioDrugs 32:299–109
    [Google Scholar]
  71. 71. 
    Kempf E, Rousseau B, Besse B, Paz-Ares L 2016. KRAS oncogene in lung cancer: focus on molecularly driven clinical trials. Eur. Respir. Rev. 25:13971–76
    [Google Scholar]
  72. 72. 
    Cheng H, Janakiram M, Borczuk A et al. 2017. HHLA2, a new immune checkpoint member of the B7 family, is widely expressed in human lung cancer and associated with EGFR mutational status. Clin. Cancer Res. 23:3825–32
    [Google Scholar]
  73. 73. 
    Sartor O, Armstrong AJ, Ahaghotu C et al. 2020. Survival of African-American and Caucasian men after sipuleucel-T immunotherapy: outcomes from the PROCEED registry. Prostate Cancer Prostatic Dis 23:517–26
    [Google Scholar]
  74. 74. 
    Guerrero S, López-Cortés A, Indacochea A et al. 2018. Analysis of racial/ethnic representation in select basic and applied cancer research studies. Sci. Rep. 8:113978
    [Google Scholar]
  75. 75. 
    Loree JM, Anand S, Dasari A et al. 2019. Disparity of race reporting and representation in clinical trials leading to cancer drug approvals from 2008 to 2018. JAMA Oncol 5:10e191870
    [Google Scholar]
  76. 76. 
    Creed JH, Berglund AE, Rounbehler RJ et al. 2020. Commercial gene expression tests for prostate cancer prognosis provide paradoxical estimates of race-specific risk. Cancer Epidemiol. Biomarkers Prev. 29:1246–53
    [Google Scholar]
  77. 77. 
    Efstathiou E, Deshpande H, George D et al. 2014. Abstract CT313: An exploratory analysis of efficacy and safety of abiraterone acetate (AA) in black patients (pts) with metastatic castration-resistant prostate cancer (mCRPC) without prior chemotherapy (ctx). Cancer Res 74:19 Suppl.CT313
    [Google Scholar]
  78. 78. 
    George DJ, Heath EI, Sartor AO et al. 2018. Abi Race: a prospective, multicenter study of black (B) and white (W) patients (pts) with metastatic castrate resistant prostate cancer (mCRPC) treated with abiraterone acetate and prednisone (AAP). J. Clin. Oncol. 36:18 Suppl.LBA5009
    [Google Scholar]
  79. 79. 
    ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consort 2020. Pan-cancer analysis of whole genomes. Nature 578:779382–93
    [Google Scholar]
  80. 80. 
    Tang M, Joensuu H, Simes RJ et al. 2019. Challenges of international oncology trial collaboration—a call to action. Br. J. Cancer 121:7515–21
    [Google Scholar]
  81. 81. 
    Lee GD, Lee SE, Oh DYet al 2017. MET exon 14 skipping mutations in lung adenocarcinoma: clinicopathologic implications and prognostic values. J. Thorac. Oncol 12:1233–46
    [Google Scholar]
/content/journals/10.1146/annurev-med-070119-120305
Loading
/content/journals/10.1146/annurev-med-070119-120305
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error