436
Views
0
CrossRef citations to date
0
Altmetric
Editorial

Bridging the gap: how do we enroll more racial-ethnic minority patients in hematological drug trials?

&
Pages 905-910 | Received 17 Jun 2023, Accepted 18 Oct 2023, Published online: 27 Oct 2023

1. Introduction

The NIH Revitalization Act of 1993 established a mandate to encourage adequate representation of racial-ethnic minorities and women in NIH-funded research [Citation1]. Since then, there have been efforts to improve recruitment of these groups in cancer clinical trials, however, these efforts have so far been insufficient in achieving widespread meaningful equity [Citation2], with most gains being realized in solid tumor oncology [Citation3]. In this editorial, we review the current state of disparities in outcomes and clinical trial enrollment in patients with hematologic malignancies, and review current and prospective strategies to address them meaningfully.

2. Where do we currently stand?

2.1. Acute myeloid leukemia (AML), Myeloproliferative Neoplasm (MPN), and myelodysplastic syndrome (MDS)

AML is the most common acute leukemia in adults and frequently has a dire prognosis. Large studies have shown that Non-Hispanic Black (NHB) and Hispanic patients have worse outcomes regardless of disease biology [Citation4,Citation5], likely related to decreased access to intensive treatment including hematopoietic cell transplantation (HCT) [Citation6,Citation7] as well as socioeconomic factors including poverty, deprivation, and structural racism [Citation8]. There are scarce data looking at disparities in MDS and MPN but existing evidence seems to point to delay in diagnosis and referral as the main drivers of worse outcomes in the former [Citation9,Citation10] and vascular and thrombotic complications in the latter [Citation11]. It is important to note that MDS has not been historically well represented as a cancer diagnosis and as such, several research efforts have limited analyses in this diagnosis area. Despite this, previous reports have shown underrepresentation of NHB in MDS clinical trials leading to FDA approval of drugs [Citation12]. Inclusion in clinical trials has shown to somewhat remediate these differences [Citation13], and while this protective effect can be mixed in some groups [Citation14], access to novel therapies and improved supportive care appear to be the main driving factors [Citation15]. Recent work has shown that in both adult and pediatric AML trials, racial-ethnic minority patients were less likely to be enrolled [Citation16,Citation17].

2.2. Acute lymphoblastic leukemia (ALL)

ALL represents the most common leukemia in children and is an emerging malignancy in adults, with recent studies demonstrating poorer outcomes particularly in Hispanic patients in the case of B-ALL [Citation18], and NHB patients in the case of T-ALL. This appears to be due as much to biological disease differences [Citation19], as to socioeconomic [Citation20,Citation21] and treatment-related factors [Citation22]. ALL is another case where clinical trial enrollment has been noted to improve prognosis of racial-ethnic minority patients. Specifically, Hispanic patients enrolled in the CALGB 10,403 trial had outcomes comparable to non-Hispanic whites (NHW) and higher rates of treatment completion [Citation23]. However, relative enrollment of Hispanic patients in this trial was lower when compared to nationwide (15% vs. 35%), which can at least be partially explained by most patients enrolled on this trial in the Midwest, a geographical region with less Hispanic population [Citation24]. This geographically driven difference in enrollment has also been noted among a large number of acute leukemia trials evaluating both therapeutics and biobanking [Citation25].

2.3. Multiple myeloma (MM)

MM is the second most common hematologic malignancy in the US, which despite several therapeutic advancements and an overall decreasing mortality, is considered incurable. NHB have more than double the incidence of MM compared to NHW (17.43 vs 7.48 per 100,000) [Citation26]. NHB also have less aggressive biological subtypes of MM but despite this, have been reported to have worse outcomes in some analyses [Citation27]. In MM, socioeconomic disparities not only play a large role [Citation28], they appear to be a major determinant of the unequal outcomes among racial-ethnic groups [Citation29]. NHB patients appear to derive the greatest benefit from inclusion in MM clinical trials, with their inclusion remediating outcome differences [Citation30]; however, both NHB and Hispanic patients remain underrepresented in clinical trials, particularly in those that evaluate novel therapeutics [Citation31].

2.4. Non-Hodgkin Lymphoma (NHL) and Hodgkin Lymphoma (HL)

Both NHL and HL constitute a diverse group of lymphoid malignancies that possess a different pathobiology and natural history. Disparities in outcomes have been documented for both HL [Citation32] and NHL [Citation33,Citation34], affecting NHB and Hispanic patients. For most subtypes of lymphoma there does not appear to be a significant biological difference, as most disparities seem to be driven by socioeconomic and environmental factors [Citation35–38], which appear to be overcome when these are accounted for [Citation39]. An exception to this is peripheral T-cell lymphomas (PTCL), where angioimmunoblastic T-cell lymphoma and extranodal NK/T cell lymphoma are much more prevalent in Hispanics, American Indian/Alaskan Native (AI/AN) and Asian/Pacific Islander (A/PI) patients, while PTCL-not otherwise specified (NOS) and adult T-Cell lymphoma/Leukemia are more common in NHB. Across these diagnoses, the racial-ethnic minority groups have worse survival as compared to NHW [Citation40–42]. Clinical trial enrollment by itself does not seem to be sufficient to fully overcome these outcome disparities, at least for HL [Citation43]. Clinical trial enrollment continues to be insufficient in NHL overall [Citation44], and particularly in PTCL, where the need seems to be most dire [Citation45,Citation46].

2.5. Hematopoietic cell transplantation (HCT) and Chimeric Antigen Receptor (CAR) T cell therapy

The use of HCT continues to increase, with the largest gains seen in the haploidentical and mismatched donor transplants [Citation47]; however, there are known barriers to access including a relative lack of donors for racial-ethnic minority populations [Citation48] as well as decreased access to transplant centers, lack of insurance or underinsurance among other socioeconomic factors [Citation7,Citation22,Citation49–53]. An interplay of these factors has been postulated as leading to worse outcomes in these populations [Citation54–56]. Inclusion of minority populations in HCT trials has been done mostly in the context of graft-versus-host disease (GVHD) prevention in haploidentical [Citation57] and mismatched unrelated donor [Citation58,Citation59] transplants. These trials have shown an improvement in rates of complications and survival, suggesting a large benefit to racial-ethnic minority patients when they are enrolled in such studies [Citation60]. Unfortunately, equitable inclusion in other trials such as transplant vs. no transplant [Citation61], treatment of GVHD [Citation62,Citation63] or infectious complications [Citation64] remains significantly suboptimal.

Chimeric antigen receptor (CAR) T-cell therapy is a novel therapeutic strategy which has shown great results in relapsed/refractory hematologic malignancies, gaining FDA approvals in MM, NHL, and B-ALL, but despite very promising response rates and curative potential there are still significant obstacles for access that underpin preexisting socioeconomic, geographical, and racial disparities [Citation65,Citation66]. This has been reflected in decreased rates of clinical trial enrollment and real-world utilization of CAR T-cell therapy, particularly for Hispanic [Citation67] and NHB [Citation68] patients.

One unique and disparate aspect of clinical trials that transcends diagnosis-specific issues but is frequently omitted from discussion is the control arm in large, randomized trials. As the therapeutic landscape changes rapidly, it may be known well before the completion of a trial that its control arm is no longer the appropriate standard of care, and since only a fraction of the trials have cross-over designs, patients may not gain access to the superior, novel therapeutic strategy. This reality hits the racial-ethnic minorities even harder as they may lose their chance to gain access to the much needed potentially lifesaving innovative options despite clinical trial participation [Citation69].

3. How can we change the status quo?

In the US, strategies to increase representation in clinical trials can be categorized as: a) policy-level efforts from regulatory agencies and sponsors; b) trial-level efforts from investigators and institutions, and c) patient-level efforts from cancer centers, patient advocacy organizations and the wider society. Some examples of potential or ongoing efforts in each of these categories are listed in .

Figure 1. Examples of patient-, trial- and policy-level potential and ongoing efforts to increase accrual of underrepresented racial-ethnic populations in clinical trials.

Figure 1. Examples of patient-, trial- and policy-level potential and ongoing efforts to increase accrual of underrepresented racial-ethnic populations in clinical trials.

3.1. Policy-level efforts

These can potentially have the biggest impact in the shortest time, given the ability for the NIH/FDA to set binding directives for the execution of clinical trials, while only reporting is mandated under current standards. Should adequate representation be mandated, this could have a huge impact on the way we recruit patients and help us reach consensus on what truly is ‘equal representation’ [Citation70]. While some recent directives have been brought forth laying out this aspiration, these are nonbinding guidelines and there is no clear understanding of the penalty of not meeting these directives [Citation71]. Legislative efforts can also have an important impact in this field, as the recently passed Clinical Treatment Act mandates Medicaid to cover clinical trial associated costs which is expected to have an outsized impact on those vulnerable populations including racial-ethnic minorities, that rely on it as their primary form of insurance [Citation72]. National Clinical Trial Networks (NCTN) and Cooperative Groups have shown the most initiative among large clinical trial sponsors; however, their efforts remain insufficient mostly due to implementation challenges [Citation2], lack of funding, and delayed surveillance/reporting [Citation73]. While pharmaceutical industry sponsors have shown tremendous interest in this issue, coordinated efforts, especially across different pharmaceutical companies are lacking.

3.2. Trial-level efforts

These have focused on making inclusion criteria for protocols more reflective of the general population so that adequate inclusion can be achieved [Citation74,Citation75], evaluating and addressing biases [Citation76,Citation77] and barriers [Citation78,Citation79] among clinical trial personnel [Citation80], improving institutional processes [Citation81] and access [Citation82], and alleviating cost/logistic burden on patients [Citation83,Citation84]. While several of these efforts have been tested, studied and reported on isolated institution level, and frequently represent expert opinions, revamping clinical trial inclusion criteria as a concerted effort can have the largest impact toward inclusive, ‘real-world’ studies, with a paradigm shift in focus from drug-centric to patient-centric clinical trials.

3.3. Patient-level efforts

These have encompassed increasing awareness about the disparities themselves, and also the availability and potential benefit of clinical trials among all patients, especially racial-ethnic minorities [Citation85,Citation86]. Other efforts have included incorporating underrepresented groups in the development of clinical trial protocols and outreach strategies [Citation87–91]. Efforts should be made across all major diagnoses, with the greatest urgency on those malignancies that are excessively overrepresented among racial-ethnic minority groups such as B-ALL, MM, and PTCL as well in trials testing novel therapeutic strategies across various diagnoses. Clinical trials remain the backbone for improving outcomes in cancer. As the number of therapeutic agents increases rapidly, it is imperative that clinical trials and the evidence they generate represent the true demographic of the real-world where these drugs are eventually utilized. The FDA implemented regulations for reporting of race/ethnicity in 2017 [Citation92]. While this has helped delineate the problem of underrepresentation in clinical trials and drug development, there is still much work that needs to be done to mitigate these disparities [Citation93]. According to the U.S. Census Bureau, the percentage of population from a race/ethnicity other than NHW will continue to increase in the upcoming years [Citation94]. Additionally, most of the population in the world is of non-European descent, such that truly realizing the benefit from cancer therapeutic advancements on a global scale will require increasing integration and improved access to healthcare among the traditionally underrepresented and underserved groups [Citation95]. Thus, the demand for cancer therapy that has been adequately evaluated across different racial-ethnic groups will only grow, and achieving this goal is not only an ethical mandate, but ultimately a necessary condition for the scientific and evidence-based practice of medicine in the 21st century. We suggest concrete strategies to address disparate enrolment in clinical trials such that this urgent issue can be brought closer to a definitive resolution, leading to the basis for equitable access to novel therapeutic advancements for all, and making clinical trials more ‘patient-centric’ rather than their current ‘drug-centric’ focus.

Declaration of interests

S Ailawadhi has had consultancy with GSK, Sanofi, BMS, Takeda, Beigene, Janssen, Regeneron, Cellectar, Pfizer and received institutional research funding from GSK, BMS, Pharmacyclics, Amgen, Janssen, Cellectar, AbbVie, Ascentage, and Sanofi.

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial relationships or otherwise to disclose.

Additional information

Funding

This paper was not funded.

References

  • S.1 - 103rd congress (1993-1994): national institutes of Health revitalization act of 1993. Congress Gov LoC. [1993 Jun 10]. https://www.congress.gov/bill/103rd-congress/senate-bill/1
  • Murthy VH, Krumholz HM, Gross CP. Participation in Cancer clinical trials. JAMA. 2004;291(22):2720. doi: 10.1001/jama.291.22.2720
  • Javier‐Desloges J, Nelson TJ, Murphy JD, et al. Disparities and trends in the participation of minorities, women, and the elderly in breast, colorectal, lung, and prostate cancer clinical trials. Cancer. 2022;128(4):770–777. doi: 10.1002/cncr.33991
  • Patel MI, Ma Y, Mitchell BS, et al. Age and genetics: how do prognostic factors at diagnosis explain disparities in acute myeloid leukemia? Am J Clin Oncol. 2015;38(2):159–164. doi: 10.1097/COC.0b013e31828d7536
  • Abraham IE, Patel AA, Wang H, et al. Impact of race on outcomes in intermediate-risk acute myeloid leukemia. Cancer Causes & Control. 2021;32(7):705–712. doi: 10.1007/s10552-021-01422-4
  • Patel MI, Ma Y, Mitchell B, et al. How do differences in treatment impact racial and ethnic disparities in acute myeloid leukemia? Cancer Epidemiol Biomarkers Prev. 2015 Feb;24(2):344–349. doi: 10.1158/1055-9965.EPI-14-0963
  • Badar T, Litzow MR, Shallis RM, et al. Disparities in receiving disease‐directed therapy, allogeneic stem cell transplantation in non‐Hispanic black patients with TP53 ‐mutated acute myeloid leukemia. Cancer. 2023;129(6):934–945. doi: 10.1002/cncr.34604
  • Abraham IE, Rauscher GH, Patel AA, et al. Structural racism is a mediator of disparities in acute myeloid leukemia outcomes. Blood. 2022 Apr 7;139(14):2212–2226. doi: 10.1182/blood.2021012830
  • Zandberg DP, Hendrick F, Vannorsdall E, et al. Tertiary center referral patterns for patients with myelodysplastic syndrome are indicative of age and race disparities: a single-institution experience. Leukemia Lymphoma. 2013;54(2):304–309. doi: 10.3109/10428194.2012.710904
  • Magalhaes SMM, Heredia FF, Fernandes LCM, et al. Beyond race-related disparities: is myelodysplastic syndrome the same everywhere? Leukemia Lymphoma. 2013;54(7):1564–1566. doi: 10.3109/10428194.2012.749404
  • Khan I, Shergill A, Saraf SL, et al. Outcome disparities in Caucasian and non-Caucasian patients with myeloproliferative neoplasms. Clin Lymphoma Myeloma Leuk. 2016 Jun;16(6):350–357. doi: 10.1016/j.clml.2016.02.036
  • Al Hadidi S, Mims M, Miller-Chism CN, et al. Participation of African American persons in clinical trials supporting U.S. Food and drug Administration approval of Cancer drugs. Ann Intern Med. 2020 Aug 18;173(4):320–322. doi: 10.7326/M20-0410
  • Buckley SA, Percival M-E, Othus M, et al. A comparison of patients with acute myeloid leukemia and high-risk myelodysplastic syndrome treated on versus off study. Leukemia Lymphoma. 2019;60(4):1023–1029. doi: 10.1080/10428194.2018.1516036
  • Truong TH, Pole JD, Barber R, et al. Enrollment on clinical trials does not improve survival for children with acute myeloid leukemia: a population‐based study. Cancer. 2018;124(20):4098–4106. doi: 10.1002/cncr.31728
  • Alexander TB, Wang L, Inaba H, et al. Decreased relapsed rate and treatment-related mortality contribute to improved outcomes for pediatric acute myeloid leukemia in successive clinical trials. Cancer. 2017;123(19):3791–3798. doi: 10.1002/cncr.30791
  • Winestone LE, Getz KD, Miller TP, et al. The role of acuity of illness at presentation in early mortality in black children with acute myeloid leukemia. Am J Hematol. 2017;92(2):141–148. doi: 10.1002/ajh.24605
  • Hantel A, Luskin MR, Garcia JS, et al. Racial and ethnic enrollment disparities and demographic reporting requirements in acute leukemia clinical trials. Blood Adv. 2021 Nov 9;5(21):4352–4360. doi: 10.1182/bloodadvances.2021005148
  • Shah N, Rockwell B, Kazemi M, et al. Ethnic disparities in survival of adult B-cell acute lymphoblastic leukemia in modern era – a SEER analysis. Leukemia Lymphoma. 2020;61(14):3503–3506. doi: 10.1080/10428194.2020.1808211
  • Lee SHR, Antillon-Klussmann F, Pei D, et al. Association of Genetic Ancestry with the molecular subtypes and prognosis of childhood acute lymphoblastic leukemia. JAMA Oncol. 2022;8(3):354. doi: 10.1001/jamaoncol.2021.6826
  • Castellanos MI, Oluyomi AO, Chambers TM, et al. Ethnic disparities in childhood leukemia survival by border residence: a Texas population‐based analysis. Cancer. 2023;129(8):1276–1286. doi: 10.1002/cncr.34636
  • Abrahão R, Lichtensztajn DY, Ribeiro RC, et al. Racial/Ethnic and socioeconomic disparities in survival among children with acute lymphoblastic leukemia in California, 1988-2011: a population-based observational study. Pediatr Blood Cancer. 2015;62(10):1819–1825. doi: 10.1002/pbc.25544
  • Jabo B, Morgan JW, Martinez ME, et al. Sociodemographic disparities in chemotherapy and hematopoietic cell transplantation utilization among adult acute lymphoblastic and acute myeloid leukemia patients. PLoS One. 2017;12(4):e0174760. doi: 10.1371/journal.pone.0174760
  • Muffly L, Yin J, Jacobson S, et al. Disparities in trial enrollment and outcomes of Hispanic adolescent and young adult acute lymphoblastic leukemia. Blood Adv. 2022 Jul 26;6(14):4085–4092. doi: 10.1182/bloodadvances.2022007197
  • Stock W, Luger SM, Advani AS, et al. A pediatric regimen for older adolescents and young adults with acute lymphoblastic leukemia: results of CALGB 10403. Blood. 2019;133(14):1548–1559. doi: 10.1182/blood-2018-10-881961
  • Hantel A, Kohlschmidt J, Eisfeld A-K, et al. Inequities in alliance acute leukemia clinical trial and biobank participation: defining targets for intervention. J Clin Oncol. 2022;40(32):3709–3718. doi: 10.1200/JCO.22.00307
  • Ellington TD, Henley SJ, Wilson RJ, et al. Trends in solitary plasmacytoma, extramedullary plasmacytoma, and plasma cell myeloma incidence and myeloma mortality by racial‐ethnic group, United States 2003‐2016. Cancer Med. 2021;10(1):386–395. doi: 10.1002/cam4.3444
  • Smith CJ, Ambs S, Landgren O. Biological determinants of health disparities in multiple myeloma. Blood Cancer J. 2018;8(9). doi: 10.1038/s41408-018-0118-z
  • Castañeda-Avila MA, Jesdale BM, Beccia A, et al. Differences in survival among multiple myeloma patients in the United States SEER population by neighborhood socioeconomic status and race/ethnicity. Cancer Causes & Control. 2021;32(9):1021–1028. doi: 10.1007/s10552-021-01454-w
  • Fillmore NR, Yellapragada SV, Ifeorah C, et al. With equal access, African American patients have superior survival compared to white patients with multiple myeloma: a VA study. Blood. 2019 Jun 13;133(24):2615–2618. doi: 10.1182/blood.2019000406
  • Aung TN, Bickell NA, Jagannath S, et al. Do patients with multiple myeloma enrolled in clinical trials live longer? Am J Clin Oncol. 2021 Dec 1;44(12):603–612. doi: 10.1097/COC.0000000000000873
  • Ailawadhi S, Jacobus S, Sexton R, et al. Disease and outcome disparities in multiple myeloma: exploring the role of race/ethnicity in the Cooperative group clinical trials. Blood Cancer J. 2018 Jul 6;8(7):67. doi: 10.1038/s41408-018-0102-7
  • Evens AM, Antillón M, Aschebrook-Kilfoy B, et al. Racial disparities in Hodgkin’s lymphoma: a comprehensive population-based analysis. Ann Oncol. 2012 Aug;23(8):2128–2137. doi: 10.1093/annonc/mdr578
  • Komrokji RS, Al Ali NH, Beg MS, et al. Outcome of diffuse large B-Cell lymphoma in the United States has improved over time but racial disparities remain: review of SEER data. Clin Lymphoma Myeloma Leuk. 2011 Jun;11(3):257–260. doi: 10.1016/j.clml.2011.03.012
  • Abodunrin FO, Akinyemi OA, Ojo AS, et al. Racial disparities in survival among non-Hodgkin lymphoma patients: an analysis of the SEER database (2007-2015). Cureus. 2022. doi:10.7759/cureus.25867
  • Vaughn JL, Spies D, Xavier AC, et al. Racial disparities in the survival of patients with indolent non-Hodgkin lymphomas in the United States. Am J Hematol. 2021;96(7):816–822. doi: 10.1002/ajh.26198
  • Wiese D, Stroup AM, Shevchenko A, et al. Disparities in cutaneous T-Cell lymphoma incidence by race/ethnicity and Area-based socioeconomic status. Int J Environ Res Public Health. 2023;20(4):3578. doi: 10.3390/ijerph20043578
  • Yung RL, Chen K, Abel GA, et al. Cancer disparities in the context of Medicaid insurance: a comparison of survival for acute myeloid leukemia and Hodgkin’s lymphoma by Medicaid enrollment. Oncology. 2011;16(8):1082–1091. doi: 10.1634/theoncologist.2011-0126
  • Shah NN, Xi Y, Liu Y, et al. Racial and socioeconomic disparities in Mantle cell Lymphoma. Clin Lymphoma Myeloma Leuk. 2019 Jun;19(6):e312–e320. doi: 10.1016/j.clml.2019.03.006
  • Williams MH, Williams RA, Blaize JP, et al. The impact of race and ethnicity on diffuse large B-Cell lymphoma (DLBCL) outcomes within the Veterans Health Administration (VHA). Blood. 2020;136(Supplement 1):3–4. doi: 10.1182/blood-2020-142118
  • Adams SV, Newcomb PA, Shustov AR. Racial patterns of peripheral T-Cell Lymphoma incidence and survival in the United States. J Clin Oncol. 2016;34(9):963–971. doi: 10.1200/JCO.2015.63.5540
  • Ai WZ, Chang ET, Fish K, et al. Racial patterns of extranodal natural killer/T-cell lymphoma, nasal type, in California: a population-based study. Br J Haematol. 2012;156(5):626–632. doi: 10.1111/j.1365-2141.2011.08982.x
  • Furqan F, Ahn KW, Chen Y, et al. Allogeneic haematopoietic cell transplant in patients with relapsed/refractory anaplastic large cell lymphoma. Br J Haematol. 2023;200(1):54–63. doi: 10.1111/bjh.18467
  • Kahn JM, Kelly KM, Pei Q, et al. Survival by race and ethnicity in pediatric and adolescent patients with Hodgkin Lymphoma: a children’s Oncology group study. J Clin Oncol. 2019;37(32):3009–3017. doi: 10.1200/JCO.19.00812
  • Gopishetty S, Kota V, Guddati AK. Age and race distribution in patients in phase III oncology clinical trials. Am J Transl Res. 2020;12(9):5977–5983.
  • Horwitz SM, Advani RH, Bartlett NL, et al. Objective responses in relapsed T-cell lymphomas with single-agent brentuximab vedotin. Blood. 2014 May 15;123(20):3095–3100. doi: 10.1182/blood-2013-12-542142
  • O’Connor OA, Pro B, Pinter-Brown L, et al. Pralatrexate in patients with relapsed or refractory peripheral T-cell lymphoma: results from the pivotal PROPEL study. J Clin Oncol. 2011 Mar 20;29(9):1182–1189. doi: 10.1200/JCO.2010.29.9024
  • National Marrow donor Program, a contractor for the C.W. Bill young cell transplantation Program operated through the U. S. Department of Health and Human Services, Health Resources and Services Administration, Healthcare Systems Bureau. Donor Registry Transplant Data. Last Updated: April 13, 2022
  • Gragert L, Eapen M, Williams E, et al. HLA match likelihoods for hematopoietic stem-cell grafts in the U.S. Registry. N Engl J Med. 2014;371(4):339–348. doi: 10.1056/NEJMsa1311707
  • Majhail NS, Nayyar S, Santibañez MEB, et al. Racial disparities in hematopoietic cell transplantation in the United States. Bone Marrow Transplant. 2012;47(11):1385–1390. doi: 10.1038/bmt.2011.214
  • Schriber JR, Hari PN, Ahn KW, et al. Hispanics have the lowest stem cell transplant utilization rate for autologous hematopoietic cell transplantation for multiple myeloma in the United States: a CIBMTR report. Cancer. 2017;123(16):3141–3149. doi: 10.1002/cncr.30747
  • Vaughn JL, Soroka O, Epperla N, et al. Racial and ethnic differences in the utilization of autologous transplantation for lymphoma in the United States. Cancer Med. 2021;10(20):7330–7338. doi: 10.1002/cam4.4249
  • Delamater PL, Uberti JP. Geographic access to hematopoietic cell transplantation services in the United States. Bone Marrow Transplant. 2016;51(2):241–248. doi: 10.1038/bmt.2015.246
  • Mock J, Meyer C, Mau LW, et al. Barriers to access to hematopoietic cell transplantation among patients with acute myeloid leukemia in Virginia. Transplant Cell Ther. 2021 Oct;27(10):e869.1–.e869.9. doi: 10.1016/j.jtct.2021.06.030
  • Bhandari R, Teh JB, He T, et al. Social vulnerability and risk of nonrelapse mortality after allogeneic hematopoietic cell transplantation. J Natl Cancer Inst. 2022 Nov 14;114(11):1484–1491. doi: 10.1093/jnci/djac150
  • Jeurkar C, Leiby B, Banks J, et al. An examination of cytomegalovirus, socioeconomic status, race, and ethnicity on outcomes after haploidentical hematopoietic transplantation. Transplant Cell Ther. 2021 Apr;27(4):.e327.1–.e327.11. doi: 10.1016/j.jtct.2020.11.017
  • Harney SM, Kahn JM, Jin Z, et al. Race and socioeconomic status in pediatric allogeneic hematopoietic cell transplantation for nonmalignant conditions. Pediatr Blood Cancer. 2020;67(9). doi: 10.1002/pbc.28367
  • Luznik L, O’Donnell PV, Symons HJ, et al. HLA-haploidentical bone marrow transplantation for hematologic malignancies using nonmyeloablative conditioning and high-dose, posttransplantation cyclophosphamide. Biol Blood Marrow Transplant. 2008 Jun;14(6):641–650. doi: 10.1016/j.bbmt.2008.03.005
  • Qayed M, Watkins B, Gillespie S, et al. Abatacept for GVHD prophylaxis can reduce racial disparities by abrogating the impact of mismatching in unrelated donor stem cell transplantation. Blood Adv. 2022 Feb 8;6(3):746–749. doi: 10.1182/bloodadvances.2021005208
  • Shaw BE, Jimenez-Jimenez AM, Burns LJ, et al. National Marrow donor Program–sponsored multicenter, phase II trial of HLA-Mismatched unrelated donor Bone Marrow transplantation using post-Transplant cyclophosphamide. J Clin Oncol. 2021;39(18):1971–1982. doi: 10.1200/JCO.20.03502
  • Thornton CP, Bandeen-Roche K, Dolinar M, et al. Clinical trial participation predicts improved survival in older adults receiving allogeneic blood and marrow transplant. BMC Geriatr. 2023;23(1). doi: 10.1186/s12877-023-03803-7
  • Nakamura R, Saber W, Martens MJ, et al. Biologic assignment trial of reduced-intensity hematopoietic cell transplantation based on donor availability in patients 50-75 years of Age with advanced myelodysplastic syndrome. J Clin Oncol. 2021;39(30):3328–3339. doi: 10.1200/JCO.20.03380
  • Zeiser R, Von Bubnoff N, Butler J, et al. Ruxolitinib for Glucocorticoid-refractory acute Graft-versus-Host disease. N Engl J Med. 2020;382(19):1800–1810. doi: 10.1056/NEJMoa1917635
  • Zeiser R, Polverelli N, Ram R, et al. Ruxolitinib for glucocorticoid-refractory chronic graft-versus-host disease. N Engl J Med. 2021;385(3):228–238. doi: 10.1056/NEJMoa2033122
  • Marty FM, Ljungman P, Chemaly RF, et al. Letermovir prophylaxis for cytomegalovirus in hematopoietic-cell transplantation. N Engl J Med. 2017;377(25):2433–2444. doi: 10.1056/NEJMoa1706640
  • Alqazaqi R, Schinke C, Thanendrarajan S, et al. Geographic and racial disparities in access to chimeric antigen receptor–t cells and bispecific antibodies trials for multiple myeloma. JAMA Netw Open. 2022;5(8):e2228877. doi: 10.1001/jamanetworkopen.2022.28877
  • Ahmed N, Shahzad M, Shippey E, et al. Socioeconomic and racial disparity in chimeric antigen receptor T cell therapy access. Transplant Cell Ther. 2022 Jul;28(7):358–364. doi: 10.1016/j.jtct.2022.04.008
  • Hall AG, Winestone LE, Sullivan EM, et al. Access to CAR T-cell clinical trials in underrepresented populations: a multicenter cohort study of pediatric and young adult ALL patients. Transplant Cell Ther. 2023 Mar 24;29(6):.e356.1–.e356.7. doi: 10.1016/j.jtct.2023.03.022
  • Al Hadidi S, Schinke C, Thanendrarajan S, et al. Enrollment of black participants in pivotal clinical trials supporting US Food and drug administration approval of chimeric antigen receptor–T cell therapy for hematological malignant neoplasms. JAMA Netw Open. 2022;5(4):e228161. doi: 10.1001/jamanetworkopen.2022.8161
  • Mohyuddin GR, Mehra N, Ryll B, et al. Control participants of randomised trials: an often forgotten, vulnerable population. Lancet Haematol. 2022 Sep;9(9):e634–e636. doi: 10.1016/S2352-3026(22)00254-X
  • Varma T, Gross CP, Miller JE. Clinical trial diversity—will we know it when we see it? JAMA Oncol. 2023;9(6):765. doi: 10.1001/jamaoncol.2023.0143
  • Diversity plans to improve enrollment of participants from underrepresented racial and ethnic populations in clinical trials guidance for industry. Rockville MD: Food and Drug Administration. 2022.
  • Karlovitch S Clinical treatment act aims to improve survival, close racial Health care gap: targeted Oncology. [cited 2021 Jun 28]. Available from: https://www.targetedonc.com/view/clinical-treatment-act-aims-to-improve-survival-close-racial-health-care-gap
  • Carpenter WR, Tyree S, Wu Y, et al. A surveillance system for monitoring, public reporting, and improving minority access to cancer clinical trials. Clin Trial. 2012;9(4):426–435. doi: 10.1177/1740774512449531
  • Riner AN, Freudenberger DC, Herremans KM, et al. Call to action: overcoming enrollment disparities in cancer clinical trials with modernized eligibility criteria. JNCI Cancer Spectr. 2023 Mar 1;7(2). doi: 10.1093/jncics/pkad009
  • Snyder RA. Clinical trial eligibility criteria: a structural barrier to diversity in clinical trial enrollment. J Clin Oncol. 2022;40(20):2183–2185. doi: 10.1200/JCO.22.00537
  • Niranjan SJ, Martin MY, Fouad MN, et al. Bias and stereotyping among research and clinical professionals: perspectives on minority recruitment for oncology clinical trials. Cancer. 2020;126(9):1958–1968. doi: 10.1002/cncr.32755
  • Niranjan SJ, Durant RW, Wenzel JA, et al. Training needs of clinical and research professionals to optimize minority recruitment and retention in Cancer clinical trials. J Cancer Educ. 2019;34(1):26–34. doi: 10.1007/s13187-017-1261-0
  • Niranjan SJ, Wenzel JA, Martin MY, et al. Perceived institutional barriers among clinical and research professionals: minority participation in Oncology clinical trials. JCO Oncology Practice. 2021;17(5):e666–e675. doi: 10.1200/OP.20.00970
  • Durant RW, Wenzel JA, Scarinci IC, et al. Perspectives on barriers and facilitators to minority recruitment for clinical trials among cancer center leaders, investigators, research staff, and referring clinicians: enhancing minority participation in clinical trials (EMPaCT). Cancer. 2014;120(S7):1097–1105. doi: 10.1002/cncr.28574
  • Penberthy L, Brown R, Wilson-Genderson M, et al. Barriers to therapeutic clinical trials enrollment: differences between African-American and white cancer patients identified at the time of eligibility assessment. Clin Trial. 2012;9(6):788–797. doi: 10.1177/1740774512458992
  • Mittal N, Davidson J, Martinez MF, et al. A tri-institutional approach to address disparities in children’s Oncology group clinical trial accrual for adolescents and young adults and underrepresented minorities. J Adolesc Young Adult Oncol. 2019 Jun;8(3):227–235. doi: 10.1089/jayao.2018.0119
  • Vicini F, Nancarrow-Tull J, Shah C, et al. Increasing accrual in cancer clinical trials with a focus on minority enrollment. Cancer. 2011;117(20):4764–4771. doi: 10.1002/cncr.26094
  • Borno HT, Zhang L, Siegel A, et al. At what cost to clinical trial enrollment? A retrospective study of patient travel burden in Cancer clinical trials. Oncology. 2018;23(10):1242–1249. doi: 10.1634/theoncologist.2017-0628
  • Ghebre RG, Jones LA, Wenzel JA, et al. State-of-the-science of patient navigation as a strategy for enhancing minority clinical trial accrual. Cancer. 2014;120(S7):1122–1130. doi: 10.1002/cncr.28570
  • Rearden J, Hanlon A, Ulrich C, et al. Examining differences in opportunity and eligibility for Cancer clinical trial participation based on sociodemographic and disease characteristics. Oncol Nurs Forum. 2016;43(1):57–66. doi: 10.1188/16.ONF.57-66
  • Leiter A, Diefenbach MA, Doucette J, et al. Clinical trial awareness: changes over time and sociodemographic disparities. Clin Trial. 2015;12(3):215–223. doi: 10.1177/1740774515571917
  • Ledesma Vicioso N, Lin D, Gomez DR, et al. Implementation strategies to increase clinical trial enrollment in a community-academic partnership and impact on Hispanic representation: an interrupted time series analysis. JCO Oncology Practice. 2022;18(5):e780–e785. doi: 10.1200/OP.22.00037
  • McClary TS, Blee SM, Avinger AM, et al. Accounting for the high enrollment of African Americans on Winship cancer institute’s myeloma clinical trials. Ethics Med Public Health. 2023 Apr; 27. doi: 10.1016/j.jemep.2023.100877
  • Wenzel JA, Mbah O, Xu J, et al. A Model of Cancer clinical trial decision-making informed by African-American Cancer patients. J Racial Ethn Health Disparities. 2015;2(2):192–199. doi: 10.1007/s40615-014-0063-x
  • Pathak S, George N, Monti D, et al. Evaluating adaptation of a Cancer clinical trial decision aid for rural Cancer patients: a mixed-methods approach. J Cancer Educ. 2019;34(4):803–809. doi: 10.1007/s13187-018-1377-x
  • Napoles A, Cook E, Ginossar T et al. Applying a conceptual framework to maximize the participation of diverse populations in Cancer clinical trials. Elsevier. 2017;133:77–94.
  • Administration USFa D FDAAA 801 and the final rule: NIH U.S. National Library of Medicine; [cited 2023]. Available from: https://clinicaltrials.gov/ct2/manage-recs/fdaaa#:~:text=Section%20801%20of%20FDAAA%20amended,and%2For%20results%20submission%20requirements
  • Jayakrishnan T, Aulakh S, Baksh M, et al. Landmark Cancer clinical trials and real-world patient populations: examining race and Age reporting. Cancers. 2021;13(22):5770. doi: 10.3390/cancers13225770
  • Bureau USC. Census results 2020. 2020. Available from: https://www.census.gov/programs-surveys/decennial-census/decade/2020/2020-census-results.html
  • Division UNDoEaSAP. World population prospects 2022. 2022. Available from: https://population.un.org/wpp/

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.