Molecular downstaging: a new paradigm for neoadjuvant endocrine therapy
Editorial Commentary

Molecular downstaging: a new paradigm for neoadjuvant endocrine therapy

Emanuela Ferraro1,2, Monica N. Fornier1,3

1Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA;2Department of Hematology and Hemato-Oncology, University of Milan, Milan, Italy;3Weill Cornell Medical College, New York, NY, USA

Correspondence to: Monica N. Fornier. Breast Medicine Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA. Email: fornierm@mskcc.org.

Comment on: Prat A, Saura C, Pascual T, et al. Ribociclib plus letrozole versus chemotherapy for postmenopausal women with hormone receptor-positive, HER2-negative, luminal B breast cancer (CORALLEEN): an open-label, multicentre, randomised, phase 2 trial. Lancet Oncol 2020;21:33-43.


Submitted Jul 20, 2020. Accepted for publication Aug 04, 2020.

doi: 10.21037/atm-20-5406


The indication for neoadjuvant endocrine therapy (NET), alone or in combination with other biological agents, is not well established. Hormone receptor (HR)-positive human epidermal growth factor receptor 2 (HER2)-negative breast cancers (BC) are generally less responsive to neoadjuvant treatments, resulting in pathological complete response (pCR) rates of 10–20% and 5% with chemotherapy (1) and endocrine therapy (2), respectively. Given the longer time needed to obtain a response from endocrine therapy compared to chemotherapy, NET was initially considered an option only for elderly patients with inoperable tumours not eligible for neoadjuvant chemotherapy (NAC) (3,4). In the last 20 years, several trials have demonstrated the benefit of NET in downstaging the primary tumour and achieving breast conserving surgery. Nevertheless, none of these studies was able to establish a final recommendation for patients’ selection and duration of treatment because underpowered, with different duration of treatment and different primary endpoints. A meta-analysis (5), published in 2016 including 3,490 patients from 20 clinical trials, compared NET to NAC and found that NET with an aromatase inhibitors (AI) yielded comparable clinical and radiological responses with similar rates of breast conservation. As expected, the toxicity was less in patients treated with NET. The metanalysis also confirmed that AI are more effective than tamoxifen. Selection criteria for NET and duration of treatment, especially if combined with biological agents, remain open questions. Dixon et al. (6) showed, that patients who received neoadjuvant letrozole beyond 3 months had a better clinical response rate compared to patients treated for 3 months only (83.5% vs. 69.8%, respectively). Given the mechanism of action of endocrine treatment, a longer duration seems to have an important role in downstaging and downsizing the tumour (7,8). The addition of CDK4/6 inhibitors to standard endocrine treatment could increase the efficacy and has opened the possibility to avoid the chemotherapy in early stage. In this article we will comment on the results of CORALEEN trial and we will review the most recent literature on NET plus CDK4/6 inhibitors.

NET gives a window of opportunity to study endocrine sensitivity in a dynamic fashion. Several endpoints have been tested to evaluate the efficacy of NET. We now have enough evidence that pCR is not a surrogate endpoint of survival for well-differentiated BC, namely luminal A-like subtype (9). This is however not the case for luminal B-like tumours, where a possible prognostic role has been reported, though not as strong as for HER2-positive and triple negative BC (9,10). The ALTERNATE study (11) presented at the 2020 ASCO meeting, which evaluated the efficacy of either anastrozole, fulvestrant or its combination in patients with stage II–III HR-positive BC, confirmed the clinical utility of Ki-67 changes as an early marker of endocrine sensitivity. Patients with Ki-67 >10% at week 4 or 12 were recommended to go off protocol-directed endocrine therapy and switch to NAC. Using this strategy, less than 2% of patients progressed during treatment. The Preoperative Endocrine Prognostic Index score (PEPI score), first tested in the P024 neoadjuvant trial and validated in the IMPACT trial (12), is largely used instead of pCR in trials evaluating NET. It combines tumour size and nodal status, as well as Ki-67 and ER expression post-NET. However, nor the change in Ki-67 or the PEPI score take in consideration the genomic features of the tumour, which have been shown to be prognostic in the early stage, and to identify the benefit of adjuvant chemotherapy.

The “Neoadjuvant Multi-Agent Chemotherapy or Letrozole plus Ribociclib in Luminal B/HER2-negative BC” (CORALLEEN trial) (13), is a randomized phase II study designed testing the hypothesis of a molecular downstaging through the PAM50 signature. Patients with high-risk luminal-B tumours were randomized to receive the CDK4/6 inhibitor ribociclib in combination with endocrine treatment for 24 weeks or standard anthracycline and taxane-containing chemotherapy. Molecular downstaging was defined as the switch from baseline high/intermediate risk-of-relapse (ROR) group, as defined by the PAM50 molecular signature, plus clinicopathological features, to a low ROR group (14). The study population included patients with stage I–IIIA BC, tumour size >2 cm and luminal-B genomic profile—the latter assessed at baseline, during treatment and at surgery. The authors aimed to validate the hypothesis that the addition of CDK4/6 to standard NET offers the possibility to induce a molecular downstaging therefore to avoid adjuvant chemotherapy in luminal B-like tumours.

The ROR risk score is the first composite biomarker which incorporates the genomic status according to the PAM50 panel. Currently, no genomic tool is approved to support the recommendation of neoadjuvant treatment and molecular signatures, such as Oncotype DX and MammaPrint, are approved in adjuvant setting (15,16).

The authors reported that among 106 patients enrolled, 87% were high ROR-risk and 16% intermediate risk at the diagnosis. The rate of conversion to low-ROR risk was similar for ribociclib and chemotherapy arms (46.9% vs. 46.1%), with a rate of pCR higher in the chemotherapy arm (5.8% vs. 0%). The conversion in low-ROR risk after 15 days of treatment occurred in 96% of cases in ribociclib arm and in only 37% in the chemotherapy arm, suggesting the potential role of CD4/6 inhibitors to induce an early molecular downstaging. While the survival data has been not collected, the authors proposed molecular downstaging as a potential biomarker of the benefit of CDK4/6 inhibitors in the neoadjuvant setting. However, the CORALLEEN study was not designed and powered for this aim.

Will the CORALLEEN trial set a new standard of care for HR-positive early BC patients? Probably not, considering that it was designed with an exploratory goal. However, this trial offers new important insight and establishes a new paradigm of study for HR-positive HER2-negative tumours—traditionally considered as poorer responders to neoadjuvant approach. This trial has underlined how important is to choose and validate the most suitable biomarkers tailored to type of tumour and class of drugs.

Previous experience with CDK4/6 inhibitors in the neoadjuvant setting have produced mixed results, with different surrogate endpoints used to evaluate efficacy and outcome (Table 1).

Table 1
Table 1 Clinical trials with CDK4/6 or biological agents in combination with endocrine treatment in neoadjuvant setting
Full table

The NeoPAL trial (17), which included patients with PAM50 luminal B-like tumour or luminal A-like with nodal involvement, compared standard chemotherapy with anthracycline and taxane vs. letrozole plus palbociclib. The experimental arm with palbociclib was associated with low rate of pCR compared to chemotherapy but encouraging responses in decreasing of Ki-67 levels as biomarker of cell proliferation. In the PALLET trial (19) with endocrine treatment alone or in combination with palbociclib, the change in the mitotic rate (Ki-67) was also used as a biomarker of response. The addition of palbociclib to letrozole enhanced significantly the suppression of cell proliferation assessed by Ki-67. Similar findings regarding decline in Ki-67 have been shown with anastrozole and/or abemaciclib in NeoMONARCH trial (21). Confirming the role in inhibiting proliferation, the NeoPalAna trial (20), with the sequential use of AI followed by palbociclib, showed a Ki-67 <2.7% observed after 15 days of the combination treatment, namely a comparative complete cell cycle arrest (CCCA)—especially in less differentiated tumours. The N007 study (22), a pivotal trial (n=20 patients) with palbociclib and letrozole administered for 16 weeks, showed a clinical response rate of 85%, with 40% of complete response. Furthermore, EndoPredict score, a multigene score that combines the expression of proliferative and ESR1 signalling/differentiation-associated genes, was reduced after treatment, suggesting a suppressive effect of the combination on proliferation pathways. Recently, the first results of FELINE trials (24) has been presented and showed no significant benefit of the addition of ribociclib to AI alone in term of PEPI score at time of surgery. The data about survival outcome have not been presented yet. Alongside trials with CD4/6 inhibitors, other targeted therapies have been investigated in this setting. For instance, in a randomized phase II neoadjuvant trial, the PIK3CA inhibitor taselisib, has been demonstrated to be more effective than letrozole alone in terms of objective response rate as assessed by magnetic resonance imaging (MRI) in the overall study patients population and PIK3A mutant cohort (23). Despite the heterogeneity of these studies in terms of different control arms, primary endpoints and duration of the treatment, the addition of biological agents seems to enhance the efficacy of endocrine treatment as monotherapy. None of these studies have been able to demonstrate the superiority of NET compared to chemotherapy and none of the biomarkers used to define the response has been validated with survival outcome data. The results of CORALEEN open new perspectives for the design of trials for early-stage luminal-like BC eligible for a neoadjuvant approach. The choice of luminal-B like population is instrumental to demonstrate that, not only in the metastatic setting but also in early stage, chemotherapy may be omitted in a subset of patients.

pCR, largely used as primary endpoint in neoadjuvant trials, seems to be a questionable predictive factor of survival outcome in patients with endocrine-sensitive tumours. The best biomarker to select patients who could avoid chemotherapy has not been well-defined yet and several questions remain unsolved regarding both surgery and medical goals. The design of future NET clinical trials should include specific endpoints to address the unmet needs in this field, considering the drugs utilized and the characteristics of the disease. Large powered studies are needed to validate the prognostic and predictive value of these endpoints. Additionally, in the evaluation of the risk of recurrence, genomic profiling of the tumour may give more comprehensive reproducible data on tumour aggressiveness compared to traditional Ki-67. The challenge is translating the precision medicine approach from the metastatic setting to the curative setting. The CORALLEEN study represents an important milestone in this direction.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the editorial office, Annals of Translational Medicine. The article did not undergo external peer review.

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/atm-20-5406). The authors have no conflicts of interests to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Prat A, Fan C, Fernández A, et al. Response and survival of breast cancer intrinsic subtypes following multi-agent neoadjuvant chemotherapy. BMC Med 2015;13:303. [Crossref] [PubMed]
  2. Ellis MJ, Suman VJ, Hoog J, et al. Randomized phase II neoadjuvant comparison between letrozole, anastrozole, and exemestane for postmenopausal women with estrogen receptor-rich stage 2 to 3 breast cancer: Clinical and biomarker outcomes and predictive value of the baseline PAM50-based intrinsic subtype - ACOSOG Z1031. J Clin Oncol 2011;29:2342-9. [Crossref] [PubMed]
  3. Mustacchi G, Ceccherini R, Milani S, et al. Italian Cooperative Group GRETA Tamoxifen alone versus adjuvant tamoxifen for operable breast cancer of the elderly: long-term results of the phase III randomized controlled multicenter GRETA trial. Ann Oncol 2003;14:414-20. [Crossref] [PubMed]
  4. Gazet JC, Ford HT, Coombes RC, et al. Prospective randomized trial of tamoxifen vs surgery in elderly patients with breast cancer. Eur J Surg Oncol 1994;20:207-14. [PubMed]
  5. Spring LM, Gupta A, Reynolds KL, et al. Neoadjuvant Endocrine Therapy for Estrogen Receptor-Positive Breast Cancer: A Systematic Review and Meta-analysis. JAMA Oncol 2016;2:1477-86. [Crossref] [PubMed]
  6. Dixon JM, Renshaw L, Macaskill EJ, et al. Increase in response rate by prolonged treatment with neoadjuvant letrozole. Breast Cancer Res Treat 2009;113:145-51. [Crossref] [PubMed]
  7. Fontein DB, Charehbili A, Nortier JW, et al. Efficacy of six month neoadjuvant endocrine therapy in postmenopausal, hormone receptor-positive breast cancer patients--a phase II trial. Eur J Cancer 2014;50:2190-200. [Crossref] [PubMed]
  8. Allevi G, Strina C, Andreis D, et al. Increased pathological complete response rate after a long-term neoadjuvant letrozole treatment in postmenopausal oestrogen and/or progesterone receptor-positive breast cancer. Br J Cancer 2013;108:1587-92. [Crossref] [PubMed]
  9. Cortazar P, Zhang L, Untch M M, et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet 2014;384:164-72. [Crossref] [PubMed]
  10. Spring LM, Fell G, Arfe A, et al. Pathological complete response after neoadjuvant chemotherapy and impact on breast cancer recurrence and survival: a comprehensive meta-analysis. Clin Cancer Res 2020. [Crossref] [PubMed]
  11. Ma CX, Suman VJ, Leitch AM, et al. ALTERNATE: Neoadjuvant endocrine treatment (NET) approaches for clinical stage II or III estrogen receptor-positive HER2-negative breast cancer (ER+ HER2- BC) in postmenopausal (PM) women: Alliance A011106. J Clin Oncol 2020;38:abstr 504.
  12. Ellis MJ, Tao Y, Luo J, et al. Outcome prediction for estrogen receptor-positive breast cancer based on postneoadjuvant endocrine therapy tumor characteristics. J Natl Cancer Inst 2008;100:1380-8. [Crossref] [PubMed]
  13. Prat A, Saura C, Pascual T, et al. Ribociclib plus letrozole versus chemotherapy for postmenopausal women with hormone receptor-positive, HER2-negative, luminal B breast cancer (CORALLEEN): an open-label, multicentre, randomised, phase 2 trial. Lancet Oncol 2020;21:33-43. [Crossref] [PubMed]
  14. Wallden B, Storhoff J, Nielsen T, et al. Development and verification of the PAM50-based Prosigna breast cancer gene signature assay. BMC Med Genomics 2015;8:54. [Crossref] [PubMed]
  15. Sparano JA, Gray RJ, Makower DF, et al. Adjuvant chemotherapy guided by a 21-gene expression assay in breast cancer. N Engl J Med 2018;379:111-21. [Crossref] [PubMed]
  16. Cardoso F, Van’t Veer LJ, Bogaerts J, et al. 70-Gene signature as an aid to treatment decisions in early-stage breast cancer. N Engl J Med 2016;375:717-29. [Crossref] [PubMed]
  17. Cottu P, D'Hondt V, Dureau S, et al. Letrozole and palbociclib versus chemotherapy as neoadjuvant therapy of high-risk luminal breast cancer. Ann Oncol 2018;29:2334-40. [Crossref] [PubMed]
  18. Ma CX, Gao F, Luo J, et al. NeoPalAna: Neoadjuvant Palbociclib, a Cyclin-Dependent Kinase 4/6 Inhibitor, and Anastrozole for Clinical Stage 2 or 3 Estrogen Receptor-Positive Breast Cancer. Clin Cancer Res 2017;23:4055-65. [Crossref] [PubMed]
  19. Johnston S, Puhalla S, Wheatley D, et al. Randomized phase II study evaluating palbociclib in addition to letrozole as neoadjuvant therapy in estrogen receptor–positive early breast cancer: Pallet trial. J Clin Oncol 2019;37:178-89. [Crossref] [PubMed]
  20. Curigliano G, Gómez Pardo P, Meric-Bernstam F, et al. Ribociclib plus letrozole in early breast cancer: A presurgical, window-of-opportunity study. Breast 2016;28:191-8. [Crossref] [PubMed]
  21. Hurvitz SA, Martin M, Press MF, et al. Potent cell-cycle inhibition and upregulation of immune response with abemaciclib and anastrozole in Neomonarch, phase II neoadjuvant study in HR+/HER2- Breast cancer. Clin Cancer Res 2020;26:566-80. [Crossref] [PubMed]
  22. Chow LWC, Morita S, Chow CYC, et al. Neoadjuvant palbociclib on ER+ breast cancer (N007): clinical response and EndoPredict's value. Endocr Relat Cancer 2018;25:123-30. [Crossref] [PubMed]
  23. Saura C, Hlauschek D, Oliveira M, et al. Neoadjuvant letrozole plus taselisib versus letrozole plus placebo in postmenopausal women with oestrogen receptor-positive, HER2-negative, early-stage breast cancer (LORELEI): a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol 2019;20:1226-38. [Crossref] [PubMed]
  24. Khan QJ, O'Dea A, Bardia A, et al. Letrozole + ribociclib versus letrozole + placebo as neoadjuvant therapy for ER+ breast cancer (FELINE trial). J Clin Oncol 2020;38:abstr 505.
Cite this article as: Ferraro E, Fornier MN. Molecular downstaging: a new paradigm for neoadjuvant endocrine therapy. Ann Transl Med 2020;8(24):1694. doi: 10.21037/atm-20-5406

Download Citation