Skip to main content

Exceptional Responders

  • Chapter
  • First Online:
Precision Cancer Medicine
  • 494 Accesses

Abstract

Anecdotes of patients who experience unexpectedly profound and/or durable responses to a specific cancer treatment, inconsistent with the experience of the vast majority of patients who receive that same treatment, are replete within the scientific literature [1]. The relevance of such outlier responses has usually not been well defined; however, in the context of clinical trials of anti-cancer therapies in which only a minority of patients (or even a single patient) achieves a substantial response, durable responses can often have significant repercussions. The efficacy of such drugs is by definition interpreted based upon the response of the overall study population, frequently leading to a failure of further drug development. Before the advent of Next Generation Sequencing (NGS), detailed analysis of patient responders was extremely challenging. Moreover, clinical trials of targeted therapies in large part initially adopted traditional histology-based eligibility criteria, leading to the accrual of patients whose tumors did not harbor the putative target inhibited by targeted agents. The phenotype of enhanced sensitivity to a drug observed within outlier cases or “exceptional responders” can now be interrogated using NGS to define their genomic underpinnings and ultimately to use this information to inform the design of future clinical trials. Additionally, outlier responders represent a unique opportunity to identify new predictive biomarkers of exquisite sensitivity to targeted therapy (or traditional chemotherapy), delineating a subset of patients that could significantly benefit from these drugs as compared to the overall population. In contrast to other strategies to identify actionable targets, such as the large-scale analyses of The Cancer Genome Atlas (TCGA) [2], examination of exceptional responders represents a phenotype-to-genotype approach in an individual, outcome-driven context that has the potential to enhance our understanding of tumor biology, inform clinical trial design, identify novel therapeutic targets, salvage drugs deemed ineffective within a genetically undefined patient population, and define more precise subgroups of patients who may benefit from new or existing drugs. In this chapter, we will describe examples of how detailed genomic analysis of extreme responses to a variety of treatments has resulted in improved insight into the molecular pathogenesis of a specific disease subtype or uncovered putative therapeutic targets for further investigation.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 84.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 109.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 109.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Similar content being viewed by others

References

  1. Chang DK, Grimmond SM, Evans TR et al (2014) Mining the genomes of exceptional responders. Nat Rev Cancer 14:291–292

    Article  CAS  Google Scholar 

  2. Weinstein JN, Collisson EA, Mills GB et al (2013) The cancer genome atlas pan-cancer analysis project. Nat Genet 45:1113–1120

    Article  Google Scholar 

  3. Iyer G, Hanrahan AJ, Milowsky MI et al (2012) Genome sequencing identifies a basis for everolimus sensitivity. Science 338:221

    Article  CAS  Google Scholar 

  4. Milowsky MI, Iyer G, Regazzi AM et al (2013) Phase II study of everolimus in metastatic urothelial cancer. BJU Int 112:462–470

    Article  CAS  Google Scholar 

  5. Lopez-Lago MA, Okada T, Murillo MM et al (2009) Loss of the tumor suppressor gene NF2, encoding merlin, constitutively activates integrin-dependent mTORC1 signaling. Mol Cell Biol 29:4235–4249

    Article  CAS  Google Scholar 

  6. James MF, Han S, Polizzano C et al (2009) NF2/merlin is a novel negative regulator of mTOR complex 1, and activation of mTORC1 is associated with meningioma and schwannoma growth. Mol Cell Biol 29:4250–4261

    Article  CAS  Google Scholar 

  7. Hudes G, Carducci M, Tomczak P et al (2007) Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med 356:2271–2281

    Article  CAS  Google Scholar 

  8. Motzer RJ, Escudier B, Oudard S et al (2010) Phase 3 trial of everolimus for metastatic renal cell carcinoma : final results and analysis of prognostic factors. Cancer 116:4256–4265

    Article  CAS  Google Scholar 

  9. Molina AM, Ginsberg MS, Motzer RJ (2011) Long-term response with everolimus for metastatic renal cell carcinoma refractory to sunitinib. Med Oncol 28:1527–1529

    Article  CAS  Google Scholar 

  10. Cheng DT, Mitchell TN, Zehir A et al (2015) Memorial sloan kettering-integrated mutation profiling of actionable cancer targets (MSK-IMPACT): a hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J Mol Diagn 17:251–264

    Article  CAS  Google Scholar 

  11. Voss MH, Hakimi AA, Pham CG et al (2014) Tumor genetic analyses of patients with metastatic renal cell carcinoma and extended benefit from mTOR inhibitor therapy. Clin Cancer Res 20:1955–1964

    Article  CAS  Google Scholar 

  12. Gerlinger M, Rowan AJ, Horswell S et al (2012) Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 366:883–892

    Article  CAS  Google Scholar 

  13. Ikezoe T, Yang Y, Nishioka C et al (2006) Effect of SU11248 on gastrointestinal stromal tumor-T1 cells: enhancement of growth inhibition via inhibition of 3-kinase/Akt/mammalian target of rapamycin signaling. Cancer Sci 97:945–951

    Article  CAS  Google Scholar 

  14. Wagle N, Grabiner BC, Van Allen EM et al (2014) Activating mTOR mutations in a patient with an extraordinary response on a phase I trial of everolimus and pazopanib. Cancer Discov 4:546–553

    Article  Google Scholar 

  15. Al-Ahmadie H, Iyer G, Hohl M et al (2014) Synthetic lethality in ATM-deficient RAD50-mutant tumors underlies outlier response to cancer therapy. Cancer Discov 4:1014–1021

    Article  CAS  Google Scholar 

  16. Peng G, Woodman SE, Mills GB (2014) RADical response puts an exceptional responder in CHKmate: a synthetic lethal curative response to DNA-damaging chemotherapy? Cancer Discov 4:988–990

    Article  CAS  Google Scholar 

  17. Frampton GM, Fichtenholtz A, Otto GA et al (2013) Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol 31:1023–1031

    Article  CAS  Google Scholar 

  18. Mehnert JM, Panda A, Zhong H et al (2016) Immune activation and response to pembrolizumab in POLE-mutant endometrial cancer. J Clin Invest 126:2334–2340

    Article  Google Scholar 

  19. Kandoth C, Schultz N, Cherniack AD et al (2013) Integrated genomic characterization of endometrial carcinoma. Nature 497:67–73

    Article  Google Scholar 

  20. van Gool IC, Eggink FA, Freeman-Mills L et al (2015) POLE proofreading mutations elicit an antitumor immune response in endometrial cancer. Clin Cancer Res 21:3347–3355

    Article  Google Scholar 

  21. Santin AD, Bellone S, Buza N et al (2016) Regression of chemotherapy-resistant polymerase epsilon (POLE) ultra-mutated and MSH6 hyper-mutated endometrial tumors with nivolumab. Clin Cancer Res 22:5682–5687

    Article  CAS  Google Scholar 

  22. Van Allen EM, Mouw KW, Kim P et al (2014) Somatic ERCC2 mutations correlate with cisplatin sensitivity in muscle-invasive urothelial carcinoma. Cancer Discov 4:1140–1153

    Article  Google Scholar 

  23. Farley J, Brady WE, Vathipadiekal V et al (2013) Selumetinib in women with recurrent low-grade serous carcinoma of the ovary or peritoneum: an open-label, single-arm, phase 2 study. Lancet Oncol 14:134–140

    Article  CAS  Google Scholar 

  24. Grisham RN, Sylvester BE, Won H et al (2015) Extreme outlier analysis identifies occult mitogen-activated protein kinase pathway mutations in patients with low-grade serous ovarian cancer. J Clin Oncol 33:4099–4105

    Article  CAS  Google Scholar 

  25. Wagle N, Grabiner BC, Van Allen EM et al (2014) Response and acquired resistance to everolimus in anaplastic thyroid cancer. N Engl J Med 371:1426–1433

    Article  Google Scholar 

  26. Wagle N, Emery C, Berger MF et al (2011) Dissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profiling. J Clin Oncol 29:3085–3096

    Article  CAS  Google Scholar 

  27. Abrams J, Conley B, Mooney M et al (2014) National cancer institute’s precision medicine initiatives for the new national clinical trials network. Am Soc Clin Oncol Educ Book 34:71–76

    Article  Google Scholar 

  28. Mullard A (2014) Learning from exceptional drug responders. Nat Rev Drug Discov 13:401–402

    Article  CAS  Google Scholar 

  29. Lawrence MS, Stojanov P, Polak P et al (2013) Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499:214–218

    Article  CAS  Google Scholar 

  30. Willyard C (2013) ‘Basket studies’ will hold intricate data for cancer drug approvals. Nat Med 19:655

    Article  CAS  Google Scholar 

  31. Redig AJ, Janne PA (2015) Basket trials and the evolution of clinical trial design in an era of genomic medicine. J Clin Oncol 33:975–977

    Article  CAS  Google Scholar 

  32. McNeil C (2015) NCI-MATCH launch highlights new trial design in precision-medicine era. J Natl Cancer Inst 107:djv193

    Article  Google Scholar 

  33. Lih CJ, Harrington RD, Sims DJ et al (2017) Analytical validation of the next-generation sequencing assay for a nationwide signal-finding clinical trial: molecular analysis for therapy choice clinical trial. J Mol Diagn 19:313–327

    Article  Google Scholar 

  34. Hyman DM, Puzanov I, Subbiah V et al (2015) Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. N Engl J Med 373:726–736

    Article  CAS  Google Scholar 

  35. Mehra N, Lorente D, de Bono JS (2015) What have we learned from exceptional tumour responses?: review and perspectives. Curr Opin Oncol 27:267–275

    Article  CAS  Google Scholar 

  36. Forshew T, Murtaza M, Parkinson C et al (2012) Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med 4:136ra68

    Article  Google Scholar 

  37. Murtaza M, Dawson SJ, Tsui DW et al (2013) Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 497:108–112

    Article  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Gopa Iyer .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2019 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Cheng, M., Iyer, G. (2019). Exceptional Responders. In: Roychowdhury, S., Van Allen, E. (eds) Precision Cancer Medicine. Springer, Cham. https://doi.org/10.1007/978-3-030-23637-3_6

Download citation

Publish with us

Policies and ethics