Can post-neoadjuvant therapy molecular classification guide future treatment selection for muscle-invasive bladder cancer?
Editorial

Can post-neoadjuvant therapy molecular classification guide future treatment selection for muscle-invasive bladder cancer?

David J. VanderWeele1,2, Joshua J. Meeks2,3, Maha Hussain1,2

1Division of Hematology/Oncology, Department of Internal Medicine, 2Robert H. Lurie Comprehensive Cancer Center, 3Department of Urology, Northwestern University, Chicago, IL, USA

Correspondence to: Maha Hussain, MD, FACP, FASCO. Robert H. Lurie Comprehensive Cancer Center, Northwestern University, 303 E. Superior St., Suite 3-107, Chicago, IL 60611, USA. Email: maha.hussain@northwestern.edu.

Provenance: This is an invited Editorial commissioned by Section Editor Xiao Li (Department of Urology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China).

Comment on: Seiler R, Gibb EA, Wang NQ, et al. Divergent biological response to neoadjuvant chemotherapy in muscle-invasive bladder cancer. Clin Cancer Res 2018. [Epub ahead of print].


Submitted Dec 04, 2018. Accepted for publication Dec 13, 2018.

doi: 10.21037/tau.2018.12.07


While platinum based chemotherapy has been the backbone of systemic therapy for muscle invasive bladder cancer for decades, little is known about how this treatment changes the tumor phenotype, or if subsequent management decisions should be based on those changes. Seiler et al. present interesting data on clustering and classification of post-chemotherapy tumors. Their data support their model that chemotherapy is not simply selecting out resistant subclones but it is also inducing changes that may promote resistance and potentially vulnerabilities for subsequent therapy.

Multiple groups have developed molecular classifications of treatment-naïve muscle invasive bladder cancers with the hope that their application to pre-treatment tumors may identify a pattern of sensitivity to guide care (1-5). At the basic level, separation of tumors into basal or luminal subtypes suggest that basal tumors may benefit the most from neoadjuvant chemotherapy (NAC), based on improved survival (but not pathologic response) (6,7). To enable implementation of this classification scheme, Seiler previously developed a single-patient classifier based on four subtypes (luminal, basal, claudin low and luminal infiltrated) (7). Yet the application of this pre-chemotherapy subtyping has not been adopted, and the intrinsic plasticity of subtypes remains controversial.

In a follow-up manuscript, Seiler applied clustering and classification to 116 matched tumors “resistant” to chemotherapy (8). While 42% of tumors remained the same subtype after NAC, 34% of tumors were in an immune infiltrated cluster (called CC3) and 12% were scar-like (called CC4). Luminal tumors were more likely to become scar-like than basal tumors (12/55 vs. 2/61, P=0.006). The CC3 immune cluster, which expresses neither luminal nor basal markers, appears to be a new subtype not seen prior to chemotherapy, with expression of immune exhaustion markers (CTLA4, MPEG1 and CD27). It appears instead that the cluster is driven by loss of differentiation markers, presumably driven by response to chemotherapy, as much as infiltrating immune cells. It is not clear if the scar-like phenotype (CC4) represents an intrinsic tumor cell phenotype or simply the phenotype of the stromal response in the setting of sensitive, though not eradicated, tumor. The authors separately analyzed scar tissue from patients with a pathologic complete response, and its expression profile is very similar to that of CC4.

Similar to the molecular subtypes of chemotherapy naïve tumors, these post-chemotherapy subtype clusters raise further questions about the instability and heterogeneity of MIBC subtypes. In particular, the immune CC3 expresses inhibitory immune genes, suggesting they might enrich for the ~20% of tumors that respond to checkpoint inhibitors. Moreover, their model suggests chemotherapy induces immune cell infiltration and an immune-responsive phenotype, perhaps converting immune depleted tumors to inflamed. It is not clear from the clinical data that prior chemotherapy enhances immune responses (9-12).

How these observations affect clinical practice remains to be seen. To contribute to management decisions in the clinic, data regarding the post-treatment consensus clusters and subsequent therapy response is needed. To be fully integrated into clinical management decisions, there is the need to develop single sample subtype classifiers, as the authors did with treatment-naïve tissue. These classifiers will be adopted only after validation from trials evaluating treatment response prospectively guided by subtype classification. Moreover, with the recent report of significant responses to neoadjuvant immunotherapy (13), there is great opportunity to develop better multitargeted combination strategies to maximize benefits for muscle invasive bladder cancer patients.


Acknowledgements

The study was supported by Prostate Cancer Foundation, Hope Foundation, VHA, NIH, DoD.


Footnote

Conflicts of Interest: JJ Meeks—Consultant: Merck, AstraZeneca, Ferring, Cold Genesys, Janssen; Research Funding: Abbvie, Tesaro, Epizyme; Compensation for talks: AUA, OncLive; Clinical Trials: SWOG, Genentech, Merck, AstraZeneca. M Hussain—Research support through contracts with Northwestern University: Genentech, AstraZeneca, Bayer; Honorarium: AstraZeneca, Pfizer, Sanofi Genzyme; Travel support: Pfizer, Sanofi Genzyme, Genentech. DJ VanderWeele has no conflicts of interest to declare.


References

  1. Sjödahl G, Lauss M, Lövgren K, et al. A Molecular Taxonomy for Urothelial Carcinoma. Clin Cancer Res 2012;18:3377-86. [Crossref] [PubMed]
  2. The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of urothelial bladder carcinoma. Nature 2014;507:315-22. [Crossref] [PubMed]
  3. Damrauer JS, Hoadley KA, Chism DD, et al. Intrinsic subtypes of high-grade bladder cancer reflect the hallmarks of breast cancer biology. Proc Natl Acad Sci U S A 2014;111:3110-5. [Crossref] [PubMed]
  4. Choi W, Porten S, Kim S, et al. Identification of distinct basal and luminal subtypes of muscle-invasive bladder cancer with different sensitivities to frontline chemotherapy. Cancer Cell 2014;25:152-65. [Crossref] [PubMed]
  5. Kardos J, Chai S, Mose LE, et al. Claudin-low bladder tumors are immune infiltrated and actively immune suppressed. JCI Insight 2016;1:e85902. [Crossref] [PubMed]
  6. Robertson AG, Kim J, Al-Ahmadie H, et al. Comprehensive Molecular Characterization of Muscle-Invasive Bladder Cancer. Cell 2017;171:540-56.e25. [Crossref] [PubMed]
  7. Seiler R, Ashab HAD, Erho N, et al. Impact of Molecular Subtypes in Muscle-invasive Bladder Cancer on Predicting Response and Survival after Neoadjuvant Chemotherapy. Eur Urol 2017;72:544-54. [Crossref] [PubMed]
  8. Seiler R, Gibb EA, Wang NQ, et al. Divergent biological response to neoadjuvant chemotherapy in muscle-invasive bladder cancer. Clin Cancer Res 2018. [Epub ahead of print]. [Crossref] [PubMed]
  9. Powles T, Durán I, van der Heijden MS, et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet 2018;391:748-57. [Crossref] [PubMed]
  10. Balar AV, Galsky MD, Rosenberg JE, et al. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet 2017;389:67-76. [Crossref] [PubMed]
  11. Balar AV, Castellano D, O’Donnell PH, et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol 2017;18:1483-92. [Crossref] [PubMed]
  12. Bellmunt J, de Wit R, Vaughn DJ, Fradet Y, et al. Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. N Engl J Med 2017;376:1015-26. [Crossref] [PubMed]
  13. Necchi A, Anichini A, Raggi D, et al. Pembrolizumab as Neoadjuvant Therapy Before Radical Cystectomy in Patients With Muscle-Invasive Urothelial Bladder Carcinoma (PURE-01): An Open-Label, Single-Arm, Phase II Study. J Clin Oncol 2018. [Epub ahead of print]. [PubMed]
Cite this article as: VanderWeele DJ, Meeks JJ, Hussain M. Can post-neoadjuvant therapy molecular classification guide future treatment selection for muscle-invasive bladder cancer? Transl Androl Urol 2019;8(Suppl 1):S91-S92. doi: 10.21037/tau.2018.12.07

Download Citation