Advances in the genomic era have led to identification of cancer-causing genes and unprecedented progress in the development of gene-targeted therapies and agents that can unleash the immune system. These advances have improved the outlook for patients with lethal malignancies.
Cancer is a genomic disease in that gene alterations drive the cellular growth and immune surveillance perturbations that enable malignant cells to take hold and to metastasize. Hereditary genetic factors play a key role in cancer predisposition, initiation, prognosis, and therapy. In a previous viewpoint, we posited that we need universal (somatic) genomic testing to win the war against cancer.1 Currently, given the rapidly emerging evidence, we update our view to state that universal germline, in addition to tumor somatic, genomic testing is needed to win the war against cancer.1
Germline genomic testing has important implications for patient diagnosis, prognosis, treatment (type of surgery, chemotherapy, radiotherapy, and novel therapeutics), screening, and for offering risk-reducing interventions, determining eligibility for clinical trials of novel agents and cascading to genetic counseling/testing of affected patients and families. Currently, most next-generation sequencing (NGS) panels involve only tumor somatic analysis. Previous guidelines for germline genome testing have been restricted to patients with known hereditary cancer syndromes thought to be at highest risk for pathogenic germline variants (PGVs) and did not favor testing all patients. Although the groups of patients indicated for germline testing has incrementally expanded, a legacy thought process remains, and stems from several perceived notions: Hereditary cancers are a rare event, high testing cost (lack of reimbursement), dearth of therapeutic options, medicolegal/ethical challenges including preexisting condition coverage, as well as lack of awareness of clinical utility, absent clear management pathways once a variant is found, discomfort in counseling patients appropriately about their risk profiles, shortage of genetic counselors, and unknown implications for approved therapies.
Currently, National Comprehensive Cancer Network recommendations include universal multigene germline testing for patients with pancreatic and ovarian cancer, as well as for those with metastatic prostate cancer2 and those younger than 50 years with colorectal cancer. For other cancers, different professional societies (and various payers) have divergent guidelines, including the genes to be tested. Importantly, restricted guideline-based testing may miss significant numbers of patients with cancer harboring germline mutations, and testing even in subsets of patients with breast cancer where guidelines are established has a low uptake in the real world, perhaps because restricted guideline-based testing leads to complexity, confusion, and practice variability. In fact, a study showed that testing all patients with breast cancer (versus guideline-based testing) doubles the number of patients identified as having an actionable germline genetic result,3 consistent with a report suggesting that restrictive testing may deny data-informed clinical management to patients with breast cancer.4 Even in other cancers with clear testing guidelines such as ovarian, pancreatic, and metastatic prostate cancers, there is significant testing underutilization, perhaps because of the complexity of guidelines between cancers.5 There is ample evidence that broad-based reflex testing of germline (providing the patient agrees) is necessary for individuals with a cancer diagnosis.
With somatic genomic alteration testing alone, we are likely missing an opportunity for screening, prevention, and offering risk reduction to close relatives of patients with cancer.6 Moreover, relatives who are not carriers need not have the apprehension about cancer risk. Furthermore, a combined tumor and germline multigene testing strategy expands our understanding of both the tumor and host. Genomics is the diagnosis, and the most powerful argument for its universal use is that every patient afflicted with cancer deserves a full diagnosis.
Where Is the Evidence?
There are multiple studies in both adults and children that support the implementation of universal genomic germline testing. In a pan-cancer study (>50 cancer types) of 11,947 patients,7 17% (n = 2,037) harbored a germline likely pathogenic or pathogenic variant; 9% (n = 1,042) had an actionable PGV.7 A prospective study of 2,984 patients with cancer examined the prevalence of PGVs using a universal germline versus target-only test on the basis of clinical guidelines, in addition to the uptake of cascade family variant testing8; one in eight patients had a PGV, one-half of which would not have been detected using a guideline-based approach. Moreover, about one-third of patients with a high-penetrance variant had revisions in their clinical management on the basis of the findings. Another study demonstrated that approximately 55% of patients with actionable PGVs would have failed to be tested under conventional guidelines.9 A recent review further showed that PGVs were found in 3.9%-56.2% of patients with common solid tumors who were unselected for family history or other putative risk factors.5 Similar data have been demonstrated in hematological malignancies as well.10 Germline mutations in cancer-predisposing genes were also identified in 8.5%-13% of multiple pan-cancer pediatric studies,11,12 and family history did not predict the underlying predisposition syndrome in most patients.11 Taken together, these data have important implications for the management of patients with cancer and their family members.
Are There Treatments?
There is an expanding armamentarium of US Food and Drug Administration (FDA)–approved precision medicines as therapeutic options, both as histology-specific and tissue-agnostic indications for many germline-aberrant cancers13: PARP inhibitors (olaparib, rucaparib, and niraparib) for treatment of germline or sporadic BRCA-mutated or homologous recombination–deficient breast, ovarian, prostate, and pancreas cancer13; immune checkpoint inhibitors (pembrolizumab and dostarlimab) for patients with germline or sporadic high microsatellite instability/mismatch repair-deficient solid tumors; nivolumab ± ipilimumab for the treatment of high microsatellite instability/mismatch repair-deficient colorectal cancers; hypoxia inducible factor 2α inhibitor belzutifan for patients with germline Von Hippel-Lindau–associated cancers; RET inhibitors for RET germline-positive medullary thyroid cancers; selumetinib for neurofibromatosis type 1 with symptomatic, inoperable plexiform neurofibromas; and everolimus for TSC1/2-associated cancers (Subependymal Giant-Cell Astrocytomas, renal angiomyolipoma and seizures).14
What About the Cost?
Many arguments against testing are centered around the cost. Yet, the cost of germline testing (and NGS in general) has markedly decreased over the past decade. Furthermore, patients get repeat routine prevention testing (mammograms, computed tomography scans for lung cancer, colonoscopies, or laboratory tests), which are quite expensive, compared with one time hereditary testing cost, which may be invaluable for the patient over an entire lifetime and informative for their family.3 In addition, the cost of the test must be weighed against the cost of expensive (and possibly futile) therapy the patient will be subjected to because this genetic information was not available in a timely manner.
What About Implementation and Legal Issues?
We acknowledge that implementation of universal genomic (somatic and germline) testing is not without challenges, including testing, coverage, reimbursement, education (including for genetic counsellors and for physicians in training and ongoing education for those in practice), and a need for more well-trained and qualified genetic counsellors.15 However, the requirement of genomic testing and services will only continue to increase, especially as universal germline testing is deployed, with the need to test and counsel family members, and the health care system needs to be ready to meet the demand, especially with access to genetic counselors who are best equipped to provide family member testing and counseling.15
As with everything in medicine and in rapidly evolving areas of science, there is no one-size-fits-all approach. Personalized medicine is focused delivering the right medicine to the right patient at the right time, and this entails the right testing for the right treatment at the right time. There are many different approaches one could envision in implementation and every clinic, hospital, health system, state, professional society, and patient advocacy group will need to find way to apply this to their population. Innovative solutions such as outlined in the ASCO educational handbook15 including telegenetics, group genetic counseling, collaboration with nongenetics heath care professionals, genetic counselor extenders, and modifications of traditional models may be required.
Because emergence of genomics has caused rapid changes to established law, the National Institutes of Health (NIH) has funded a project LawSeqSM: Building a Sound Legal Foundation for Translating Genomics into Clinical Application.16 This project seeks to address the legal challenges including how the law of liability for all stakeholders from health care professionals to industry should adjust to meet the challenge of genomics. Since the law underlying application science is ambiguous, the LawSeqSM project has convened a national Working Group of top legal and scientific experts to compile, collect, and analyze current US federal and state law and regulation on translational genomics including liability from failure to test.17-19
Current restrictive guideline-based testing misses many patients with germline alterations, especially in underserved populations (with African-American, Asian/Pacific Islander, and Hispanic populations) typically underrepresented in germline testing20 which in turn denies data-driven care to patients and amplifies disparities. A recent review of 84,297 oncology clinical trials from the Trialtrove database revealed that 887 (1.1%) trials used germline data for inclusion/exclusion, and most trials using germline data were conducted in the United States, Canada, and Europe versus other countries, mirroring disparities in cancer genomics data globally.21,22 In addition, immune checkpoint inhibitor pembrolizumab is US FDA-approved for solid tumors with high tumor mutational burden (TMB-high; ≥10 mutations/megabase) or microsatellite instability (which itself leads to a high TMB).23 It was shown recently that TMB is affected by an individual's genetic ancestry and race.24 Tumor-only sequencing overestimates TMBs especially in people of non-European ancestry, exacerbating disparities in precision medicine.24 Calibration of tumor-only TMB using paired tumor/normal TMB and/or algorithmic strategies may improve ancestral biases.25
Universal germline testing may not only transform the outlook for those with traditional hereditary cancer syndromes but also identify a wider range of associations and penetrance for germline variants. If we are serious about winning the war against cancer, we need to have every bit of intelligence about it, both to treat cancer and to detect it early. The potential impact of the host should be considered in every patient with cancer. Applying universal germline testing to patients with cancer routinely is one of the major opportunities that can revolutionize precision medicine practice and is needed to win the war against cancer (and possibly many other diseases).
ACKNOWLEDGMENT
V.S. is an Andrew Sabin Family Foundation Fellow at the University of Texas MD Anderson Cancer Center. V.S. acknowledges the support of the Jacquelyn A. Brady Fund.
Vivek Subbiah
Consulting or Advisory Role: MedImmune, Helsinn Therapeutics, Relay Therapeutics, Pfizer, Loxo/Lilly
Research Funding: Novartis (Inst), GlaxoSmithKline (Inst), NanoCarrier (Inst), Northwest Biotherapeutics (Inst), Genentech/Roche (Inst), Berg Pharma (Inst), Bayer (Inst), Incyte (Inst), Fujifilm (Inst), PharmaMar (Inst), D3 Oncology Solutions (Inst), Pfizer (Inst), Amgen (Inst), AbbVie (Inst), Multivir (Inst), Blueprint Medicines (Inst), LOXO (Inst), Vegenics (Inst), Takeda (Inst), Alfasigma (Inst), Agensys (Inst), Idera (Inst), Boston Biomedical (Inst), Inhibrx (Inst), Exelixis (Inst), Turning Point Therapeutics (Inst), Relay Therapeutics (Inst)
Travel, Accommodations, Expenses: PharmaMar, Bayer, Novartis, Helsinn Therapeutics, Foundation Medicine
Other Relationship: Medscape
Razelle Kurzrock
Leadership: CureMatch, CureMetrix
Stock and Other Ownership Interests: CureMatch, IDbyDNA, CureMetrix
Honoraria: Roche, EUSA Pharma, NeoGenomics Laboratories, Biocom, NeoMed, LEK, AACR, Chugai Pharma USA, Wiley, Merck, Pfizer, Foundation Medicine, Turning Point Therapeutics, Bicara Therapeutics
Consulting or Advisory Role: Actuate Therapeutics, Loxo, XBiotech, NeoMed, Roche, Gaido, Soluventis, Pfizer, Merck, Turning Point Therapeutics, TD2/Volastra, Bicara Therapeutics, AstraZeneca, Biological Dynamics, Daiichi Sankyo, Eisai, EOM Pharmaceuticals, Iylon, Prosperdtx
Speakers' Bureau: Roche, NeoGenomics Laboratories
Research Funding: Guardant Health (Inst), Sequenom (Inst), Merck Serono (Inst), Genentech (Inst), Pfizer (Inst), Foundation Medicine (Inst), Incyte (Inst), Konica Minolta (Inst), Grifols (Inst), OmniSeq (Inst), Debiopharm Group (Inst), Boehringer Ingelheim (Inst), Top Alliance BioScience (Inst), Takeda (Inst), MedImmune (Inst), Biological Dynamics (Inst)
Travel, Accommodations, Expenses: Target Cancer, NCI SWOG
Other Relationship: Dr Kurzrock receives research funding from Boehringer Ingelheim, Debiopharm, Foundation Medicine, Genentech, Grifols, Guardant, Incyte, Konica Minolta, Medimmune, Merck Serono, Omniseq, Pfizer, Sequenom, Takeda, and TopAlliance; as well as consultant and/or speaker fees and/or advisory board for Actuate Therapeutics, AstraZeneca, Bicara Therapeutics, Inc, Biological Dynamics, Caris, Datar Genomics, EISAI, EOM Pharmaceuticals, Iylon, Merck, NeoGenomics, Neomed, Pfizer, Prosperdtx, Regeneron, Roche, TD2/Volastra, Turning Point Therapeutics, X-Biotech; has an equity interest in CureMatch Inc and IDbyDNA; serves on the Board of CureMatch and CureMetrix, and is a co-founder of CureMatch
No other potential conflicts of interest were reported.
SUPPORT
V.S. is supported by a US National Institutes of Health (NIH; grant No. R01CA242845 and R01CA273168). The MD Anderson Cancer Center Department of Investigational Cancer Therapeutics is supported by the Cancer Prevention and Research Institute of Texas (RP1100584), the Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy (1U01 CA180964), NCATS Grant UL1 TR000371 (Center for Clinical and Translational Sciences), and the MD Anderson Cancer Center Support Grant (P30 CA016672).
AUTHOR CONTRIBUTIONS
Financial support: Vivek Subbiah
Administrative support: Vivek Subbiah
Provision of study materials or patients: Vivek Subbiah
Manuscript writing: All authors
Final approval of manuscript: All authors
Accountable for all aspects of the work: All authors
AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
Universal Germline and Tumor Genomic Testing Needed to Win the War Against Cancer: Genomics Is the Diagnosis
The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/jco/authors/author-center.
Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments).
Vivek Subbiah
Consulting or Advisory Role: MedImmune, Helsinn Therapeutics, Relay Therapeutics, Pfizer, Loxo/Lilly
Research Funding: Novartis (Inst), GlaxoSmithKline (Inst), NanoCarrier (Inst), Northwest Biotherapeutics (Inst), Genentech/Roche (Inst), Berg Pharma (Inst), Bayer (Inst), Incyte (Inst), Fujifilm (Inst), PharmaMar (Inst), D3 Oncology Solutions (Inst), Pfizer (Inst), Amgen (Inst), AbbVie (Inst), Multivir (Inst), Blueprint Medicines (Inst), LOXO (Inst), Vegenics (Inst), Takeda (Inst), Alfasigma (Inst), Agensys (Inst), Idera (Inst), Boston Biomedical (Inst), Inhibrx (Inst), Exelixis (Inst), Turning Point Therapeutics (Inst), Relay Therapeutics (Inst)
Travel, Accommodations, Expenses: PharmaMar, Bayer, Novartis, Helsinn Therapeutics, Foundation Medicine
Other Relationship: Medscape
Razelle Kurzrock
Leadership: CureMatch, CureMetrix
Stock and Other Ownership Interests: CureMatch, IDbyDNA, CureMetrix
Honoraria: Roche, EUSA Pharma, NeoGenomics Laboratories, Biocom, NeoMed, LEK, AACR, Chugai Pharma USA, Wiley, Merck, Pfizer, Foundation Medicine, Turning Point Therapeutics, Bicara Therapeutics
Consulting or Advisory Role: Actuate Therapeutics, Loxo, XBiotech, NeoMed, Roche, Gaido, Soluventis, Pfizer, Merck, Turning Point Therapeutics, TD2/Volastra, Bicara Therapeutics, AstraZeneca, Biological Dynamics, Daiichi Sankyo, Eisai, EOM Pharmaceuticals, Iylon, Prosperdtx
Speakers' Bureau: Roche, NeoGenomics Laboratories
Research Funding: Guardant Health (Inst), Sequenom (Inst), Merck Serono (Inst), Genentech (Inst), Pfizer (Inst), Foundation Medicine (Inst), Incyte (Inst), Konica Minolta (Inst), Grifols (Inst), OmniSeq (Inst), Debiopharm Group (Inst), Boehringer Ingelheim (Inst), Top Alliance BioScience (Inst), Takeda (Inst), MedImmune (Inst), Biological Dynamics (Inst)
Travel, Accommodations, Expenses: Target Cancer, NCI SWOG
Other Relationship: Dr Kurzrock receives research funding from Boehringer Ingelheim, Debiopharm, Foundation Medicine, Genentech, Grifols, Guardant, Incyte, Konica Minolta, Medimmune, Merck Serono, Omniseq, Pfizer, Sequenom, Takeda, and TopAlliance; as well as consultant and/or speaker fees and/or advisory board for Actuate Therapeutics, AstraZeneca, Bicara Therapeutics, Inc, Biological Dynamics, Caris, Datar Genomics, EISAI, EOM Pharmaceuticals, Iylon, Merck, NeoGenomics, Neomed, Pfizer, Prosperdtx, Regeneron, Roche, TD2/Volastra, Turning Point Therapeutics, X-Biotech; has an equity interest in CureMatch Inc and IDbyDNA; serves on the Board of CureMatch and CureMetrix, and is a co-founder of CureMatch
No other potential conflicts of interest were reported.
REFERENCES
- 1.Subbiah V, Kurzrock R: Universal genomic testing needed to win the war against cancer: Genomics is the diagnosis. JAMA Oncol 2:719-720, 2016 [DOI] [PubMed] [Google Scholar]
- 2.Daly MB, Pal T, Berry MP, et al. : Genetic/familial high-risk assessment: Breast, ovarian, and pancreatic, version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Cancer Netw 19:77-102, 2021 [DOI] [PubMed] [Google Scholar]
- 3.Beitsch PD, Whitworth PW, Hughes K, et al. : Underdiagnosis of hereditary breast cancer: Are genetic testing guidelines a tool or an obstacle? J Clin Oncol 37:453-460, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Whitworth PW, Beitsch PD, Patel R, et al. : Clinical utility of universal germline genetic testing for patients with breast cancer. JAMA Netw Open 5:e2232787, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Esplin ED, Nielsen SM, Bristow SL, et al. : Universal germline genetic testing for hereditary cancer syndromes in patients with solid tumor cancer. JCO Precis Oncol 6:e2100516, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Henson JW: Paired tumor-germline testing as a driver in better cancer care. JAMA Netw Open 5:e2213077, 2022 [DOI] [PubMed] [Google Scholar]
- 7.Stadler ZK, Maio A, Chakravarty D, et al. : Therapeutic implications of germline testing in patients with advanced cancers. J Clin Oncol 39:2698-2709, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Samadder NJ, Riegert-Johnson D, Boardman L, et al. : Comparison of universal genetic testing vs guideline-directed targeted testing for patients with hereditary cancer syndrome. JAMA Oncol 7:230-237, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Mandelker D, Zhang L, Kemel Y, et al. : Mutation detection in patients with advanced cancer by universal sequencing of cancer-related genes in tumor and normal DNA vs guideline-based germline testing. JAMA 318:825-835, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kraft IL, Godley LA: Identifying potential germline variants from sequencing hematopoietic malignancies. Hematology 2020:219-227, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Zhang J, Walsh MF, Wu G, et al. : Germline mutations in predisposition genes in pediatric cancer. N Engl J Med 373:2336-2346, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Fiala EM, Jayakumaran G, Mauguen A, et al. : Prospective pan-cancer germline testing using MSK-IMPACT informs clinical translation in 751 patients with pediatric solid tumors. Nat Cancer 2:357-365, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Hasanov E, Pimentel I, Cruellas M, et al. : Current systemic treatments for the hereditary cancer syndromes: drug development in light of genomic defects. Am Soc Clin Oncol Ed Book 42 1-17, 2022 [DOI] [PubMed] [Google Scholar]
- 14.Subbiah V, Wirth LJ, Kurzrock R, et al. : Accelerated approvals hit the target in precision oncology. Nat Med 28:1976-1979, 2022 [DOI] [PubMed] [Google Scholar]
- 15.Cohen SA, Bradbury A, Henderson V, et al. : Genetic counseling and testing in a community setting: quality, access, and efficiency. Am Soc Clin Oncol Ed Book 39:e34-e44, 2019 [DOI] [PubMed] [Google Scholar]
- 16.Genomics Law, 2023. https://lawseq.umn.edu/ [Google Scholar]
- 17.Marchant G, Barnes M, Evans JP, et al. : From genetics to genomics: Facing the liability implications in clinical care. J Law Med Ethics 48:11-43, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Cheung FY, Clatch L, Wolf SM, et al. : Key expert stakeholder perceptions of the law of genomics: Identified problems and potential solutions. J Law Med Ethics 48:87-104, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.LawSeq℠ : Building a Sound Legal Foundation for Translating Genomics into Clinical Application. 2023. https://consortium.umn.edu/research/genetics-genomics/lawseqsm-building-sound-legal-foundation-translating-genomics-clinical-application/4771 [Google Scholar]
- 20.Weise N, Shaya J, Javier-Desloges J, et al. : Disparities in germline testing among racial minorities with prostate cancer. Prostate Cancer Prostatic Dis 25:403-410, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Kammula AV, Schäffer AA, Rajagopal PS: Characterization of oncology clinical trials using germline genetic data. JAMA Netw Open 5:e2242370, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Moyers JT, Subbiah V: Think globally, act locally: Globalizing precision oncology. Cancer Discov 12:886-888, 2022 [DOI] [PubMed] [Google Scholar]
- 23.Subbiah V, Solit DB, Chan TA, et al. : The FDA approval of pembrolizumab for adult and pediatric patients with tumor mutational burden (TMB) ≥10: A decision centered on empowering patients and their physicians. Ann Oncol 31:1115-1118, 2020 [DOI] [PubMed] [Google Scholar]
- 24.Nassar AH, Adib E, Abou Alaiwi S, et al. : Ancestry-driven recalibration of tumor mutational burden and disparate clinical outcomes in response to immune checkpoint inhibitors. Cancer Cell 40:1161-1172.e5, 2022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Cheng C, Amos CI: A refined use of mutations to guide immunotherapy decisions. Nature 612:639-641, 2022 [DOI] [PubMed] [Google Scholar]