Review
Pseudoprogression and hyperprogression after checkpoint blockade

https://doi.org/10.1016/j.intimp.2018.03.018Get rights and content

Highlights

  • Pseudoprogression or hyperprogression could emerge after checkpoint blockade.

  • Pseudoprogression is a radiological progression followed by tumor shrinkage

  • IrRC or irRECIST have the advantage in identifying pseudoprogression.

  • Hyperprogression is a phenomenon reflecting extraordinarily rapid tumor progression.

  • The basis, identification and predictors of hyperprogression are largely unknown.

Abstract

Immune checkpoint inhibitors appear to be one of the most promising immunotherapies with significant clinical benefits and durable responses in multiple tumor types. A heterogeneity of responses appears in patients receiving checkpoint blockade, including pseudoprogression where the tumor burden or number of tumor lesions increases initially before decreasing. Another special response observed after checkpoint blockade is hyperprogression, a phenomenon reflecting a very rapid tumor progression following immunotherapy, suggesting that checkpoint blockade could impact detrimentally on a small subset of patients. As immunotherapeutics, especially anti-PD-1/PD-L1 agents, become more widely available, evaluating the efficacy of these novel drugs poses a major challenge to clinicians, who aim to avoid either premature withdrawal of the treatment or prolonging ineffective treatment. Although the mechanism and recognition of pseudoprogression have gradually come to light, the incidence, basis, identification and predictive biomarkers of hyperprogression have been largely unknown, and this review documents the existing research findings and points out the areas where further studies are badly needed.

Introduction

Immunotherapy that reestablishes antitumor response within the host is rising as a promising treatment strategy for cancer patients [[1], [2], [3]]. Immune checkpoint inhibitors, such as monoclonal antibodies(mAb) targeting cytotoxic T-lymphocyte associated antigen-4 (CTLA-4) and programmed death-1 (PD-1), are profoundly changing cancer patient management [[4], [5], [6], [7]]. At the forefront of this novel class of antitumor agents, anti-PD-1/PD-L1 antibodies exhibit a significant activity and induce durable disease control by restoring an efficient antitumor T-cell response. It has become a standard of care for multiple cancer types including melanoma, non-small-cell lung cancer (NSCLC), renal-cell carcinoma (RCC), bladder cancer, Hodgkin's lymphoma, head and neck squamous cell carcinoma (HNSCC) and Merkel cell carcinoma [[8], [9], [10], [11]]. Overall, the response rates for single agent of PD-1/PD-L1 inhibitors in solid malignancies range from 20% to 40% [[12], [13], [14]]. Till now, five immune checkpoint inhibitors targeting PD-1 and PD-L1 have been approved by the U.S. Food and Drug Administration (FDA), including PD-1-blocking mAbs pembrolizumab and nivolumab, and PD-L1-targeted mAbs atezolizumab, durvalumab and avelumab. Apart from CTLA-4 and PD-1, multiple inhibitory receptors, such as T-cell immunoglobulin mucin 3 (TIM3), lymphocyte activation gene 3 (LAG3) and B and T lymphocyte attenuator (BTLA), also compose the family of immune checkpoint and transmit similar inhibitory functions, and, hence, call for further exploration [15,16].

As immunotherapeutics, particularly anti-PD-1/PD-L1 agents, are more widely available to patients, clinicians face a great challenge in accurately evaluating the clinical efficacy of these novel drugs. WHO and RECIST criteria have historically been taken as standard guidelines in defining a tumor response to therapy. Although imperfect, the RECIST are commonly accepted norms defining the moment of disease progression and have guided the defining of tumor response and driven consequent drug approval for years [17,18]. The development of new lesions and a significant increase in the size of tumor lesions are considered definite disease progression by RECIST criteria. However, during checkpoint blockade some patients experienced immune-related responses such as initial increased size of tumor lesions or appearance of new lesions, confirmed by biopsy as necrosis or inflammatory cell infiltrates, with subsequent reduction in tumor burden. The unconventional clinical response is recognized as pseudoprogression, and would be misclassified as progressive disease (PD) according to the size-based WHO or RECIST criteria.

In addition to pseudoprogression, there is another special response pattern during checkpoint blockade named tumor flare or hyperprogression. Unlike pseudoprogression, a disease progression subsequently followed by tumor shrinkage, hyperprogression is characterized as the disease whose dramatic progression outpaces the expected rate of growth in the absence of checkpoint inhibitors, based primarily on evidence from prior imaging scans (Fig. 1). The patients with disease hyperprogression have a course that is more deleterious than they might have had with other therapies, or even in the absence of therapy. Although the mechanism and recognition of pseudoprogression have gradually come to light, the incidence, basis, identification and predictive biomarkers of hyperprogression have been largely unknown. As immunotherapeutics, especially anti-PD-1/PD-L1 agents, become more widely available, the discernment in differentiating pseudoprogression from real progression or even hyperprogression by clinicians will tremendously help evaluate the efficacy of these novel drugs and avoid either premature withdrawal of the treatment or prolonging ineffective treatment. Therefore, this review focuses on the existing researches in related areas and clarifies potential topics for future studies.

Section snippets

The occurrence of pseudoprogression after immune checkpoint blockade

The concept of “tumor pseudoprogression” was first introduced in brain tumors treated with non-immune therapy agent, temozolomide, which is a prodrug that works through alkylation (methylation) of DNA. It is discovered that brain tumors might increase in size before responding to temozolomide treatment [20]. Pseudoprogression in immune checkpoint inhibitor therapy was initially found in melanoma, at first in the research of the anti-CTLA4 inhibitor ipilimumab [21] and then in the studies of

The occurrence of hyperprogression after PD-1/PD-L1 blockade

Opposing effects of immunotherapy have already been documented in melanoma using adjuvant IFN-α where patients in the treatment group who died during the study period displayed a significantly reduced time from relapse to death compared with control individuals [60]. Anti-PD-1 and anti-PD-L1 monotherapy can lead to profound and durable tumor responses in some cases, however, the opposing effects of immunotherapy probably exist in a subset of patients. The phase III study of nivolumab versus

Conclusions

As immune checkpoint inhibitors become more widely accepted by physicians, the task of properly assessing tumor response remains a challenge in patient management. The irRC, though with some limitations such as measurement variability, may help to identify a subgroup of patients with pseudoprogression so as to avoid inappropriate discontinuations of immunotherapy. As for hyperprogression, a dramatic tumor surge in a fraction of patients after checkpoint blockade, exploration of the underlying

Acknowledgments

This work is supported by grants from National Natural Science Foundation of China (81472843). We would also like to show our gratitude to the “anonymous” reviewers for their insights that greatly improved the manuscript.

References (99)

  • L. Calabro et al.

    Efficacy and safety of an intensified schedule of tremelimumab for chemotherapy-resistant malignant mesothelioma: an open-label, single-arm, phase 2 study

    Lancet Respir. Med.

    (2015)
  • H.L. Kaufman et al.

    Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial

    Lancet Oncol.

    (2016)
  • M. Nishino et al.

    Cancer immunotherapy and immune-related response assessment: the role of radiologists in the new arena of cancer treatment

    Eur. J. Radiol.

    (2015)
  • S.S. Baxi et al.

    Amidst the excitement: a cautionary tale of immunotherapy, pseudoprogression and head and neck squamous cell carcinoma

    Oral Oncol.

    (2016)
  • R. Solana et al.

    Innate immunosenescence: effect of aging on cells and receptors of the innate immune system in humans

    Semin. Immunol.

    (2012)
  • F.T. Hakim et al.

    Aging, immunity and cancer

    Curr. Opin. Immunol.

    (2004)
  • C.K. Lee et al.

    Checkpoint inhibitors in metastatic EGFR-mutated non-small cell lung cancer-a meta-analysis

    J. Thorac. Oncol.

    (2017)
  • C. Schindler et al.

    JAK-STAT signaling: from interferons to cytokines

    J. Biol. Chem.

    (2007)
  • H.A. Tawbi et al.

    Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): a multicentre, two-cohort, single-arm, open-label, phase 2 trial

    Lancet Oncol.

    (2017)
  • S. Kleffel et al.

    Melanoma cell-intrinsic PD-1 receptor functions promote tumor growth

    Cell

    (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)
  • M. Collin

    Immune checkpoint inhibitors: a patent review (2010–2015)

    Expert Opin. Ther. Pat.

    (2016)
  • C. Massard et al.

    Safety and efficacy of durvalumab (MEDI4736), an anti-programmed cell death ligand-1 immune checkpoint inhibitor, in patients with advanced urothelial bladder cancer

    J. Clin. Oncol.

    (2016)
  • S.M. Ansell et al.

    PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma

    N. Engl. J. Med.

    (2015)
  • O. Hamid et al.

    Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma

    N. Engl. J. Med.

    (2013)
  • T. Powles et al.

    MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer

    Nature

    (2014)
  • P. Sharma et al.

    The future of immune checkpoint therapy

    Science

    (2015)
  • H. Borghaei et al.

    Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer

    N. Engl. J. Med.

    (2015)
  • C. Robert et al.

    Nivolumab in previously untreated melanoma without BRAF mutation

    N. Engl. J. Med.

    (2015)
  • S.J. Antonia et al.

    Immunotherapy: beyond anti-PD-1 and anti-PD-L1 therapies

    Am. Soc. Clin. Oncol. Educ. Book

    (2016)
  • A.E. Vilgelm et al.

    Combinatorial approach to cancer immunotherapy: strength in numbers

    J. Leukoc. Biol.

    (2016)
  • P. Therasse et al.

    New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada

    J. Natl. Cancer Inst.

    (2000)
  • M. Nishino et al.

    Revised RECIST guideline version 1.1: what oncologists want to know and what radiologists need to know

    AJR Am. J. Roentgenol.

    (2010)
  • C. Tsien et al.

    Parametric response map as an imaging biomarker to distinguish progression from pseudoprogression in high-grade glioma

    J. Clin. Oncol.

    (2010)
  • J.D. Wolchok et al.

    The mechanism of anti-CTLA-4 activity and the negative regulation of T-cell activation

    Oncologist

    (2008)
  • V.L. Chiou et al.

    Pseudoprogression and immune-related response in solid tumors

    J. Clin. Oncol.

    (2015)
  • F.S. Hodi et al.

    CTLA-4 blockade with ipilimumab induces significant clinical benefit in a female with melanoma metastases to the CNS

    Nat. Clin. Pract. Oncol.

    (2008)
  • M. Millward et al.

    Phase I study of tremelimumab (CP-675 206) plus PF-3512676 (CPG 7909) in patients with melanoma or advanced solid tumours

    Br. J. Cancer

    (2013)
  • A. Ribas et al.

    Safety profile and pharmacokinetic analyses of the anti-CTLA4 antibody tremelimumab administered as a one hour infusion

    J. Transl. Med.

    (2012)
  • Y.K. Chae et al.

    Pseudoprogression in microsatellite instability-high colorectal cancer during treatment with combination T cell mediated immunotherapy: a case report and literature review

    Oncotarget

    (2017)
  • J.D. Wolchok et al.

    Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria

    Clin. Cancer Res.

    (2009)
  • R. Danielli et al.

    Ipilimumab in pretreated patients with metastatic uveal melanoma: safety and clinical efficacy

    Cancer Immunol. Immunother.

    (2012)
  • A.M. Di Giacomo et al.

    Ipilimumab experience in heavily pretreated patients with melanoma in an expanded access program at the University Hospital of Siena (Italy)

    Cancer Immunol. Immunother.

    (2011)
  • F.S. Hodi et al.

    Evaluation of immune-related response criteria and RECIST v1.1 in patients with advanced melanoma treated with pembrolizumab

    J. Clin. Oncol.

    (2016)
  • M. Nishino et al.

    Immune-related tumor response dynamics in melanoma patients treated with pembrolizumab: identifying markers for clinical outcome and treatment decisions

    Clin. Cancer Res.

    (2017)
  • S. Atrash et al.

    Response of metastatic mucosal melanoma to immunotherapy: it can get worse before it gets better

    J. Oncol. Pharm. Pract.

    (2017)
  • F.S. Hodi et al.

    Long-term survival of ipilimumab-naive patients (pts) with advanced melanoma (MEL) treated with nivolumab (anti-PD-1, BMS-936558, ONO-4538) in a phase I trial

    J. Clin. Oncol.

    (2014)
  • S.L. Topalian et al.

    Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab

    J. Clin. Oncol.

    (2014)
  • Cited by (117)

    View all citing articles on Scopus
    View full text