Elsevier

Cancer Treatment Reviews

Volume 48, July 2016, Pages 61-68
Cancer Treatment Reviews

Tumour Review
Tumor genotype and immune microenvironment in POLE-ultramutated and MSI-hypermutated Endometrial Cancers: New candidates for checkpoint blockade immunotherapy?

https://doi.org/10.1016/j.ctrv.2016.06.008Get rights and content

Highlights

  • Survival of advanced EC is a challenge and new treatment modalities are needed.

  • EC is an immunogenic tumor, and immunotherapy might be a promising option.

  • Genotype studies showed different immune microenvironment in EC subtypes.

  • Blocking PD-1/PD-L1 axis could reduce the cancer-induced immunosuppression.

  • POLE-ultramutated, and MSI-hypermutated might most benefit from immunotherapy.

Abstract

Endometrial Cancer (EC) is still a challenge for gynecological oncologists because the treatment of the advanced disease remains an unmet need for patients. The Cancer Genome Atlas Research Network (TCGA) recently provided a comprehensive genomic and transcriptomic analysis of EC, offering a new classification of the disease, based on genetic features, which defines four subgroups of cancer rather than the two traditionally recognized. In the molecular classification two types of EC, the polymerase epsilon (POLE)-ultramutated and the microsatellite instability (MSI)-hypermutated, seem to present an enhanced immune microenvironment and a high mutation burden. The blockade of the immune checkpoints is an innovative approach that has largely demonstrated to be effective in solid malignancies, such as lung, renal and melanoma; it acts by reducing the cancer-induced immune-suppression through inhibition of the PD-1/PD-L1 (Programmed Death and PD-Ligand) axis. All available evidence supporting an over-expression of the PD-1/PD-L1 pathway in EC has been reviewed. In particular in the POLE and MSI ECs an up-regulation of this pathway was found, aiming to suggest a rationale for testing the PD-1/PD-L1 immunotherapy in these cancer subgroups.

Introduction

EC is the most common gynecological malignancy in the Western world, accounting approximately for 150,000 women per year in Europe and the United States combined [1], while the localized disease is largely curable with surgery, in some cases followed by radiotherapy [2], the treatment of the advanced EC remains an unmet need for patients. In metastatic and relapsed EC, chemotherapy and hormonal therapy are the only available options for treatment but survival remains poor [2].

With regards to chemotherapy the carboplatin–paclitaxel doublet has progressively replaced the combination of paclitaxel, adriamycine, and cisplatin in the first line setting, being equally effective but less toxic [2]. Among the hormonal agents currently used in clinical practice the most popular are progestins (megestrol acetate and medroxyprogesterone), followed by anti-estrogens (tamoxifen) and aromatase inhibitors with response rates up to 30% [2].

Nevertheless the options in second and third line are limited and the approach is absolutely not standardized, different treatments have failed to demonstrate a benefit in this setting [2]. Many new-targeted therapies have been tested in clinical trials but only few agents have shown an impact on survival and response and to date none has been approved [2]. Among them, the mTOR inhibitors had the highest response rate [3], particularly the combination of everolimus plus letrozole resulted in an objective response rate of 32% [4]. Also the combination with temsirolimus and bevacizumab was efficacious even if more toxic [5]. Bevacizumab and sunitinib as single agents have resulted in objective response rates of 12–15% [6], [7]. Use of multitargeted VEGF/FGFR inhibitors (brivatinib, lenvatinib) has produced encouraging findings (response rates 14–19%) [3], [8]. In a phase II trial, AEZS-108, an LHRH agonist conjugated to doxorubicin, was active and well-tolerated in LHRH receptor positive recurrent EC [9].

In such context new treatment modalities are urgently needed and the comprehension of the EC genotype is likely to represent a milestone in the development of new therapeutic strategies.

The mapping of the genomic landscape of ECs has recently identified 4 molecular subgroups: (1) POLE-ultramutated, (2) MSI-hypermutated (3) copy-number low and (4) copy-number high, serous like [10].

The POLE-ultramutated and the MSI groups are characterized by an active immune microenvironment demonstrated by the abundance of tumor specific neo-antigens and the high number of Tumor Infiltrating Lymphocytes (TILs) [11], leading to over-expression of PD-1 and PD-L1 [11]. Immune checkpoints other than PD-1 and PD-L1, such as CTLA-4 (Cytotoxic T Lymphocyte Antigen-4), LAG-3 (Lymphocyte Activation Gene-3), and IDO (indoleamine 2,3-dioxygenase), may also be up-regulated in the POLE and in the MSI EC, this phenomenon is known as ‘adaptive immune resistance’ [11], [12], [13].

On note, the response to the checkpoint inhibitors seems to correlate both with the number of the predicted antigenic tumor mutations and the T-cell infiltration [14], [15], [16], suggesting that patients with POLE-ultra mutated and MSI EC may have the maximum benefit from these drugs [2].

This review focuses on the available data supporting (i) the enhanced immune microenvironment and the significant mutation burden in the POLE-ultramutated and in the MSI-hypermutated EC (ii) the correlation between the efficacy of the PD-1/PD-L1 blockade and these cancer features. The aim of this review is to suggest a rational for testing the anti- PD-1/PD-L1 agents in these subgroups of EC.

Section snippets

2.1 Type I and Type II

Historically, ECs have been classified into Type I and II, as defined by Bokhman et al., on the basis of clinical, endocrine and epidemiological characteristics [17]. Subsequently, histological types and molecular features became integral components into such dualistic model (Table 1) [18]. Type I tumors, 60–70% of all ECs, present grade 1–2 endometrioid histology, hormone receptor positivity (Estrogen receptors (ERs), and Progesteron Receptors (PgRs)), history of unopposed estrogen exposure

3.1 Role of the immune system in EC

There is a large body of evidence supporting that the tumor immune microenvironment is unique and complex, it may have a crucial role in promoting carcinogenesis and maintaining malignant cells growth [47], [48], [49]. The importance of intact immune surveillance in controlling outgrowth of neoplastic transformation has been widely recognized [48], [50], [51], [52]. There are many publications demonstrating that a high level of the TILs is associated with a favorable prognosis in different

Challenges of immunotherapy in EC

Over the past decade, cancer immunotherapy has made a remarkable progress from bench to bedside. The inhibition of PD-1/PD-L1 axis has lead to clinically relevant results in different cancer types, however, only a subset of patients will likely benefit from such treatments.

Major questions regarding immunotherapy in EC patients are: who should be treated? How to integrate immunotherapy with other therapies and how to combine various immunotherapies?

Currently, immunotherapy may represent a

Conclusions

The systematic analysis of the cancer genome has generated a new scenario for the comprehension of the carcinogenesis and hopefully for cancer treatment; however, although it represents a very fascinating opportunity, we are still far from fully interpret this huge amount of information and translating them into clinical practice.

Given the high rate of PD-1/PD-L1 expression in the uterine carcinoma (up to 80%) and the already demonstrated efficacy of the PD-L1/PD-1 inhibition in many cancer

Conflict of interest

The authors declare that they have no conflicts of interest.

Funding

None.

Author contribution

PG and SP conceived and designed the manuscript; PG acquired, analyzed and interpreted data, and drafted the article; all authors revised it critically for important intellectual content, and final approved the version to be submitted.

Acknowledgment

None.

References (81)

  • I. Diaz-Padilla et al.

    Mismatch repair status and clinical outcome in endometrial cancer: a systematic review and meta-analysis

    Crit Rev Oncol Hematol

    (2013)
  • D. Hanahan et al.

    Hallmarks of cancer: the next generation

    Cell

    (2011)
  • D. Mittal et al.

    New insights into cancer immunoediting and its three component phases – elimination, equilibrium and escape

    Curr Opin Immunol

    (2014)
  • R.A. de Jong et al.

    Presence of tumor-infiltrating lymphocytes is an independent prognostic factor in type I and II endometrial cancer

    Gynecol Oncol

    (2009)
  • A. Vanderstraeten et al.

    The immune system in the normal endometrium and implications for endometrial cancer development

    J Reprod Immunol

    (2015)
  • K.A. Sheppard et al.

    PD-1 inhibits T-cell receptor induced phosphorylation of the ZAP70/CD3zeta signalosome and downstream signaling to PKCtheta

    FEBS Lett

    (2004)
  • J. Liu et al.

    Plasma cells from multiple myeloma patients express B7–H1 (PD-L1) and increase expression after stimulation with IFN-{gamma} and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway

    Blood

    (2007)
  • D.H. Munn et al.

    Indoleamine 2,3 dioxygenase and metabolic control of immune responses

    Trends Immunol

    (2013)
  • X. Meng et al.

    Predictive biomarkers in PD-1/PD-L1 checkpoint blockade immunotherapy

    Cancer Treat Rev

    (2015)
  • Y.R. Hussein et al.

    Clinicopathological analysis of endometrial carcinomas harboring somatic POLE exonuclease domain mutations

    Mod Pathol

    (2015)
  • P. Morice et al.

    Endometrial cancer

    Lancet

    (2015)
  • J.L. Silva et al.

    Endometrial cancer: redefining the molecular-targeted approach

    Cancer Chemother Pharmacol

    (2015)
  • B.M. Slomovitz et al.

    Phase II study of everolimus and letrozole in patients with recurrent endometrial carcinoma

    J Clin Oncol

    (2015)
  • K.J. Dedes et al.

    Emerging therapeutic targets in endometrial cancer

    Nat Rev Clin Oncol

    (2011)
  • M. Le Gallo et al.

    The emerging genomic landscape of endometrial cancer

    Clin Chem

    (2014)
  • B.E. Howitt et al.

    Association of polymerase e-mutated and microsatellite-instable endometrial cancers with neoantigen load, number of tumor-infiltrating lymphocytes, and expression of PD-1 and PD-L1

    JAMA Oncol

    (2015)
  • J.R. Brahmer et al.

    Safety and activity of anti-PD-L1 antibody in patients with advanced cancer

    N Engl J Med

    (2012)
  • S.L. Topalian et al.

    Safety, activity, and immune correlates of anti-PD-1 antibody in cancer

    N Engl J Med

    (2012)
  • D.M. Pardoll

    The blockade of immune checkpoints in cancer immunotherapy

    Nat Rev Cancer

    (2012)
  • N.A. Rizvi et al.

    Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer

    Science

    (2015)
  • A. Snyder et al.

    Genetic basis for clinical response to CTLA-4 blockade in melanoma

    N Engl J Med

    (2014)
  • L.W. Cheung et al.

    High frequency of PIK3R1 and PIK3R2 mutations in endometrial cancer elucidates a novel mechanism for regulation of PTEN protein stability

    Cancer Discov

    (2011)
  • M.P. Hayes et al.

    PIK3CA and PTEN mutations in uterine endometrioid carcinoma and complex atypical hyperplasia

    Clin Cancer Res

    (2006)
  • G.L. Mutter et al.

    Altered PTEN expression as a diagnostic marker for the earliest endometrial precancers

    J Natl Cancer Inst

    (2000)
  • K. Oda et al.

    High frequency of coexistent mutations of PIK3CA and PTEN genes in endometrial carcinoma

    Cancer Res

    (2005)
  • M.L. Rudd et al.

    A unique spectrum of somatic PIK3CA (p110alpha) mutations within primary endometrial carcinomas

    Clin Cancer Res

    (2011)
  • S.F. Lax et al.

    The frequency of p53, K-ras mutations, and microsatellite instability differs in uterine endometrioid and serous carcinoma: evidence of distinct molecular genetic pathways

    Cancer

    (2000)
  • H.B. Salvesen et al.

    Methylation of hMLH1 in a population-based series of endometrial carcinomas

    Clin Cancer Res

    (2000)
  • S.B. Simpkins et al.

    MLH1 promoter methylation and gene silencing is the primary cause of microsatellite instability in sporadic endometrial cancers

    Hum Mol Genet

    (1999)
  • A.J. O’Hara et al.

    The genomics and genetics of endometrial cancer

    Adv Genomics Genet

    (2012)
  • Cited by (0)

    View full text