82 results on '"Anti pd 1"'
Search Results
2. M2 subtype tumor associated macrophages (M2-TAMs) infiltration predicts poor response rate of immune checkpoint inhibitors treatment for prostate cancer
- Author
-
Wang HongYi, Liu Yang, Huang Zhi-Peng, Xia Ming, Dou ChunXia, Guo WenBing, Liao DeYing, Yang Jiankun, Xue Kangyi, Liang ZhiJian, Duan HaiFeng, Liu Cundong, Zhao ShanChao, Yang Cheng, Zhou Junhao, Chen Mingkun, Zhou Qizhao, Bao JiMing, Xie Xiao, and Zhou JiaWei
- Subjects
Male ,anti-PD-L1 ,animal diseases ,medicine.medical_treatment ,Immune checkpoint inhibitors ,Immune microenvironment ,chemical and pharmacologic phenomena ,urologic and male genital diseases ,Prostate cancer ,Immune system ,Tumor-Associated Macrophages ,medicine ,M2-TAMs ,Tumor Microenvironment ,Humans ,Immune Checkpoint Inhibitors ,Response rate (survey) ,business.industry ,Anti pd 1 ,Prostatic Neoplasms ,General Medicine ,Immunotherapy ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,Oncology ,Cancer research ,bacteria ,immune subtype ,business ,tumour microenvironment ,Infiltration (medical) ,Research Article - Abstract
Background Prostate cancer (PCa) is poor response to the immunotherapy for its high heterogeneity of immune microenvironment. In this study, we aim to introduce a new immune subtype for PCa involving M2 tumour associated macrophages (M2-TAMs). Methods Three hundred and sixty-two PCa patients and matched normal prostate tissues were selected from the Cancer Genome Atlas and Gene Expression Omnibus databases. Patients’ immune infiltration characters were then analyzed based on the gene expressions. The immune subtypes were identified by the method of unsupervised hierarchical clustering. Finally, the relationship between the M2-TAMs infiltration and anti-programmed death-ligand-1 (PD-L1) therapy was investigated in the IMvigor210 cohort. Results PCa expressed lower immune-related genes levels compared with the adjacent normal tissues. Based on the proved immunosuppressive mechanisms in PCa, tumour patients were classified into three independent subclasses with high infiltrated cytolytic activity (CYT), M2-TAMs and regulatory T cell (Tregs), respectively. Among these subtypes, M2-TAMs infiltration subtype showed the worst clinicopathological features and prognosis compared with the other two subtypes. The results of the IMvigor210 cohort demonstrated poor response of anti-PD-L1 therapy for patients with high M2-TAMs infiltration. Conclusion Prostate tumours involved in significant immunosuppression, and high infiltration of M2-TAMs can be applied to predict the effect of anti-PD-L1 therapy.Key MessagesPCa patients can be classified into three immunotypes of high infiltrated CYT, M2-TAMS, and Tregs according to the immunosuppressive mechanisms.High M2-TAMs infiltration subtype reflected the worst clinical characters, immune infiltration, and lowest expression of immune checkpoint inhibitors among the three subclasses in PCa.High M2-TAMs infiltration predicts the low response rate of anti-PD-L1 therapy.
- Published
- 2021
3. First-line BRAF/MEK inhibitors versus anti-PD-1 monotherapy in BRAF(V600)-mutant advanced melanoma patients: a propensity-matched survival analysis
- Author
-
Alfonsus J. M. van den Eertwegh, Michel W.J.M. Wouters, Roos S. van Rijn, John B. A. G. Haanen, Maureen J.B. Aarts, Bert-Jan J. ten Tije, Karijn P M Suijkerbuijk, Christian U. Blank, Jesper van Breeschoten, Geke A. P. Hospers, Jan-Willem B de Groot, Franchette W P J van den Berkmortel, Ellen Kapiteijn, Doranne L. Hilarius, Marye J Boers-Sonderen, Djura Piersma, Art Vreugdenhil, Astrid A M van der Veldt, Willeke A. M. Blokx, VU University medical center, Obstetrics and gynaecology, Internal medicine, AII - Cancer immunology, CCA - Cancer Treatment and quality of life, Radiology & Nuclear Medicine, RS: GROW - R3 - Innovative Cancer Diagnostics & Therapy, Interne Geneeskunde, and MUMC+: MA Medische Oncologie (9)
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,PHASE-3 ,First line ,Systemic therapy ,VEMURAFENIB ,Article ,Healthcare improvement science Radboud Institute for Health Sciences [Radboudumc 18] ,POOLED ANALYSIS ,Matched cohort ,SDG 3 - Good Health and Well-being ,Internal medicine ,Medicine ,METASTATIC MELANOMA ,neoplasms ,Melanoma ,Survival analysis ,Advanced melanoma ,nivolumab ,business.industry ,IPILIMUMAB ,Anti pd 1 ,medicine.disease ,DABRAFENIB ,Propensity score matching ,pembrolizumab ,business - Abstract
Contains fulltext : 232046.pdf (Publisher’s version ) (Closed access) BACKGROUND: Anti-PD-1 antibodies and BRAF/MEK inhibitors are the two main groups of systemic therapy in the treatment of BRAF(V600)-mutant advanced melanoma. Until now, data are inconclusive on which therapy to use as first-line treatment. The aim of this study was to use propensity score matching to compare first-line anti-PD-1 monotherapy vs. BRAF/MEK inhibitors in advanced BRAF(V600)-mutant melanoma patients. METHODS: We selected patients diagnosed between 2014 and 2017 with advanced melanoma and a known BRAF(V600)-mutation treated with first-line BRAF/MEK inhibitors or anti-PD-1 antibodies, registered in the Dutch Melanoma Treatment Registry. Patients were matched based on their propensity scores using the nearest neighbour and the optimal matching method. RESULTS: Between 2014 and 2017, a total of 330 and 254 advanced melanoma patients received BRAF/MEK inhibitors and anti-PD-1 monotherapy as first-line systemic therapy. In the matched cohort, patients receiving anti-PD-1 antibodies as a first-line treatment had a higher median and 2-year overall survival compared to patients treated with first-line BRAF/MEK inhibitors, 42.3 months (95% CI: 37.3-NE) vs. 19.8 months (95% CI: 16.7-24.3) and 65.4% (95% CI: 58.1-73.6) vs. 41.7% (95% CI: 34.2-51.0). CONCLUSIONS: Our data suggest that in the matched BRAF(V600)-mutant advanced melanoma patients, anti-PD-1 monotherapy is the preferred first-line treatment in patients with relatively favourable patient and tumour characteristics.
- Published
- 2021
- Full Text
- View/download PDF
4. Correction: First-line BRAF/MEK inhibitors versus anti-PD-1 monotherapy in BRAFV600-mutant advanced melanoma patients: a propensity-matched survival analysis
- Author
-
Djura Piersma, Marye J Boers-Sonderen, Geke A. P. Hospers, Karijn P M Suijkerbuijk, Christian U. Blank, Roos S. van Rijn, Astrid A M van der Veldt, Jan-Willem B de Groot, Ellen Kapiteijn, Franchette W P J van den Berkmortel, Alfonsus J. M. van den Eertwegh, Jesper van Breeschoten, Doranne L. Hilarius, Bert-Jan J. ten Tije, Michel W.J.M. Wouters, Art Vreugdenhil, John B. A. G. Haanen, Maureen J.B. Aarts, and Willeke A. M. Blokx
- Subjects
Oncology ,medicine.medical_specialty ,Cancer Research ,business.industry ,First line ,Anti pd 1 ,Mutant ,Correction ,Cancer ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,Medicine ,business ,Survival analysis ,Advanced melanoma - Abstract
A Correction to this paper has been published: https://doi.org/10.1038/s41416-021-01312-1
- Published
- 2022
- Full Text
- View/download PDF
5. Limited Impact of Anti-PD-1/PD-L1 Monotherapy for Hepatocellular Carcinoma
- Author
-
Masatoshi Kudo
- Subjects
atezolizumab ,Hepatology ,biology ,Bevacizumab ,business.industry ,Immune checkpoint inhibitors ,Anti pd 1 ,bevacizumab ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,immune checkpoint inhibitors ,Editorial ,Oncology ,Atezolizumab ,Hepatocellular carcinoma ,PD-L1 ,combination immunotherapy ,Cancer research ,medicine ,biology.protein ,Combination immunotherapy ,business ,medicine.drug - Published
- 2020
6. Consilium Smartphone App for Real-World Electronically Captured Patient-Reported Outcome Monitoring in Cancer Patients Undergoing anti-PD-L1-Directed Treatment
- Author
-
Mathis Brauchbar, Andreas Trojan, Ulf Petrausch, and Urs Huber
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Remote patient monitoring ,Case Report ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,In patient ,Cancer ,Dry cough ,business.industry ,Anti pd 1 ,Common Terminology Criteria for Adverse Events ,Usability ,Smartphone app ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,030104 developmental biology ,Oncology ,Electronically captured patient-reported outcomes ,030220 oncology & carcinogenesis ,Patient-reported outcome ,Immunotherapy ,business - Abstract
Digital patient monitoring gains importance for quality of clinical cancer care. Our case report provides insight into usability and acceptance of a smartphone app for monitoring of electronically captured patient-reported outcomes in patients undergoing immunotherapy. During 3 months, 6 patients with advanced or metastatic PD-L1-positive cancer of the lung, prostate, and bladder who underwent checkpoint immunotherapy were using the Consilium app for standardized and structured electronic reporting of symptoms and therapy side effects. We evaluated the number and quality of symptom entries as well as usability and safety of shared reporting between the patient and the treating physician. Duration of anti-PD-L1-directed immunotherapy in the 6 patients ranged from 4 to 10 months and comprised a total of 21 anti-PD-L1-directed immunotherapy cycles. Patients reported between 4 and 16 different symptoms, of which the most frequent (57%) were dry cough, fatigue, shortness of breath, fever, and appetite loss. Overall, 1,279 symptom entries were counted, corresponding to 2.4 symptom entries per patient per day. Symptom severity grading ranged from 0.1 (very slight symptoms) to 7.8 (severe symptoms), which triggered prespecified alerts in 4 of the 6 patients. No unplanned visits were noted, and no safety issues occurred. Satisfaction with the app usability was high, as was the beneficial effect on consultation. Usability and reviewed data entries indicate high shared reporting efforts of patients and treating physicians and overall satisfaction with electronically reported patient outcomes.
- Published
- 2020
7. Is there a role for neoadjuvant anti-PD-1 therapies in glioma?
- Author
-
Thomas J Lai, Robert M. Prins, and Lu Sun
- Subjects
Oncology ,Surgical resection ,medicine.medical_specialty ,medicine.medical_treatment ,Article ,Immune system ,Internal medicine ,Glioma ,Tumor Microenvironment ,Medicine ,Humans ,Tumor microenvironment ,business.industry ,Brain Neoplasms ,Anti pd 1 ,Immunotherapy ,medicine.disease ,Neoadjuvant Therapy ,Neurology ,Neurology (clinical) ,business ,Glioblastoma ,Checkpoint Blockade Immunotherapy - Abstract
PURPOSE OF REVIEW In this review, we summarized recent findings that highlight the progress for checkpoint blockade immunotherapy in glioblastoma (GBM) patients. RECENT FINDINGS We reviewed new data from our group and others that suggest that the timing of when immunotherapy is applied can impact the antitumor immune response and, potentially, the ultimate clinical benefit of patients. SUMMARY The neoadjuvant priming and expansion of exhausted T cells within the GBM microenvironment, followed by the removal of an immune suppressive tumor microenvironment through surgical resection, may lead to enhanced antitumor immune responses that are beneficial clinically. As such, neoadjuvant immunotherapeutic approaches and rational combinations may be helpful scientifically to understand how immunotherapeutic interventions influence the tumor microenvironment, as well benefit the patients.
- Published
- 2021
8. Suspected Immune-Related Adverse Events With an Anti-PD-1 Inhibitor in Otherwise Healthy People With HIV
- Author
-
William David Hardy, Steven Hendrickx, Randall Tressler, Edgar T. Overton, Joseph J. Eron, Kendall F. Moseley, Maureen Furlong, Elizabeth Miller, Michael Messer, Constance A. Benson, Arrow trial team, Chanelle L Wimbish, Daniel R. Kuritzkes, Bernard J.C. Macatangay, Cheryl Jennings, Ronald J. Bosch, Danielle M Campbell, and Ashley McKahnn
- Subjects
business.industry ,Anti pd 1 ,Human immunodeficiency virus (HIV) ,MEDLINE ,medicine.disease_cause ,Infectious Diseases ,Text mining ,Immune system ,Immunology ,medicine ,Pharmacology (medical) ,business ,Adverse effect ,Letters to the Editor - Published
- 2021
9. Anti-programmed cell death protein 1-induced lichenoid changes of the nail unit: Histopathologic description
- Author
-
Claire van Damme, Josette André, Vincent Sibaud, Bertrand Richert, and Evelyne Berlingin
- Subjects
medicine.medical_specialty ,Immune checkpoint inhibitors ,medicine.medical_treatment ,lichenoid changes ,Case Report ,Dermatology ,Programmed cell death 1 ,medicine ,lcsh:Dermatology ,nail ,Anti-PD-1, Anti-programmed cell death protein 1 ,Dermatologie ,nivolumab ,biology ,lichen planus ,business.industry ,Anti pd 1 ,Immunotherapy ,lichen unguis ,lcsh:RL1-803 ,irAEs, immune-related adverse events ,medicine.anatomical_structure ,checkpoint inhibitor ,biology.protein ,Cancer research ,Nail (anatomy) ,histopathology ,anti-PD-1 ,Histopathology ,immunotherapy ,Nivolumab ,business - Abstract
SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2021
10. 286 Sex differences in the transcriptional profiles of mucosal-associated invariant T cells in neoadjuvant anti-PD-1 treated non-small cell lung cancer (NSCLC)
- Author
-
Frank Housseau, Poromendro Burman, Justina X. Caushi, Kellie N. Smith, Hongkai Ji, Zhang Jiajia, Andrew Pardoll, Zhicheng Ji, and Boyang Zhang
- Subjects
Pharmacology ,Cancer Research ,business.industry ,Immunology ,Anti pd 1 ,non-small cell lung cancer (NSCLC) ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Mucosal associated invariant T cell ,medicine.disease ,Oncology ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,business ,RC254-282 - Abstract
BackgroundMucosal Associated Invariant T Cells (MAIT cells) are unconventional T cells that recognize vitamin B metabolites derived from bacteria and are mainly present in mucosal tissues and peripheral blood.1 Their activation by T Cell Receptor (TCR)-dependent and -independent pathways can result in effector function that can either promote or inhibit cytotoxic effects.2 MAIT cells are known to be involved in the pathogenesis of multiple diseases that involve mucosal tissues, such as non-small cell lung cancer (NSCLC).2 Recently, studies have shown that disparate outcomes to SARS-CoV-2-infection between males and females may involve a differential activation of MAIT cells in the lung mucosa.3 It is therefore conceivable to hypothesize that sex differences of MAIT cells in NSCLC may also impact outcome, however their involvement in progression and subsequent treatment response of NSCLC has never been explored.MethodsTo study the transcriptional program of MAIT cells in NSCLC as a function of sex, peripheral blood and tissue biospecimens were obtained from the first-in-human clinical trial of neoadjuvant anti-PD-1 (nivolumab) in resectable non-small cell lung cancer; NCT02259621.4 Coupled single-cell RNAseq/TCRseq was performed on tumor infiltrating lymphocytes (TIL), paired adjacent normal lung, and tumor-draining lymph nodes (TDLN). MAIT cells were identified by expression of SLC4A10 and the invariant TRAV1-2 and TRAJ33/12/20 TCR. Computational analysis revealed 4 distinct MAIT cell clusters and differentially expressed genes in the tumors and healthy normal lung of males as compared to females.ResultsIn MAIT cells from females, we found upregulation of CD8A, GNLY, and NKG7 genes. These genes are involved with T cell activation and cytolytic function, suggesting that the activation of these genes in MAIT cells could be contributing towards their cytolytic activity in females. In MAIT cells from males, we found upregulation of PDE3B and PCBP2 genes, which are known to be involved with immunosuppression and downregulation of cytotoxic T lymphocyte (CTL) responses. These findings were consistent in the healthy normal lung, suggesting these transcriptional programs may be due to the normal lung biology and not necessarily a byproduct of carcinogenesis.ConclusionsThese results highlight the potential for dual characteristics of MAIT cells in neoadjuvant anti-PD-1-treated NSCLCs and provide an important foundation in our study of the often dichotomous responses between males and females to immunotherapy. Future analyses will focus on the interplay of MAIT cells with other cells in the tumor microenvironment (TME) as a function of immunotherapy treatment and clinical response.ReferencesChen Z, Wang H, D’Souza C, et al. Mucosal-associated invariant T-cell activation and accumulation after in vivo infection depends on microbial riboflavin synthesis and co-stimulatory signals. Mucosal Immunol 2017;10:58–68.Wen X, Zhang X, et al. Title of article: mucosal-associated invariant T cells in lung cancers. Elsevier 2021;94.Yu C, Littleton S, et al. Mucosal-associated invariant T cell responses differ by sex in COVID-19. CellPress 2021;2:755–772.Caushi JX, Zhang J, Ji Z, et al. Transcriptional programs of neoantigen-specific TIL in anti-PD-1-treated lung cancers. Nature 2021.Ethics ApprovalThis study was approved by the Institutional Review Boards (IRB) at Johns Hopkins University (JHU) and Memorial Sloan Kettering Cancer Center and was conducted in accordance with the Declaration of Helsinki and the International Conference on Harmonization Good Clinical Practice guidelines. The patients described in this study provided written informed consent.
- Published
- 2021
11. 535 A phase I/II study of REGN7075 (EGFRxCD28 costimulatory bispecific antibody) in combination with cemiplimab (anti–PD-1) in patients with advanced solid tumors
- Author
-
Melissa Lynne Johnson, Frank Seebach, Dimitris Skokos, Roman Groisberg, Israel Lowy, Kerry Casey, Melissa Mathias, Siyu Li, Matthew G. Fury, Hyunsil Han, and Nehal Lakhani
- Subjects
Pharmacology ,Cancer Research ,Bispecific antibody ,Chemistry ,Immunology ,Anti pd 1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Phase i ii ,Oncology ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,In patient ,RC254-282 - Abstract
BackgroundT-cell redirecting bispecific antibodies (bsAbs) are therapeutics that recognize two distinct antigens: a tumor-associated antigen on tumor cells to promote recruitment of T-cells to the tumor, and a receptor on T-cells to potentiate anti-tumor activity. REGN7075 is a human immunoglobulin G4-based costimulatory bsAb designed to bridge epidermal growth factor receptor (EGFR) positive tumor cells with CD28 positive T-cells and to provide amplified T-cell receptor-CD3 complex-mediated T-cell activation within the tumor, through the activation of CD28 co-stimulation. In genetically humanized immunocompetent mouse models, REGN7075 in combination with anti–PD-1 (antibody directed against programmed cell death-1 receptor) improved anti-tumor activity compared with either single agent alone.1MethodsThis is an open label, Phase I/II, first-in-human study evaluating the safety, tolerability, pharmacokinetics, and preliminary anti-tumor activity of REGN7075 (EGFRxCD28) alone and in combination with cemiplimab in patients with advanced solid tumors (NCT04626635). Patients must have a protocol-defined advanced solid tumor, be ≥18 years of age (≥20 years in Japan), have an Eastern Cooperative Oncology Group performance status of 0 or 1, and be naïve to anti–PD-1/anti–PD-ligand(L)1.This study includes dose escalation (a 4+3 design modified from 3+3; Part 1) and expansion phases (Part 2). In Part 1, patients will receive a lead-in of REGN7075 monotherapy for 3 weeks followed by combination therapy with cemiplimab 350 mg every 3 weeks. Study therapies are administered until disease progression, intolerable adverse events, withdrawal of consent, or other stopping criterion is met. Once a recommended Phase 2 dose is determined in Part 1, four tumor-specific expansion cohorts will be opened: non-small cell lung cancer (PD-L1 ≥50%), triple-negative breast cancer, colorectal cancer (microsatellite stable), and cutaneous squamous cell carcinoma. Primary endpoints are safety and tolerability of REGN7075 alone or in combination with cemiplimab for Part 1, and objective response rate per Response Evaluation Criteria in Solid Tumors version 1.1 for Part 2. This study is currently open to enrollment.Trial RegistrationClinicalTrials.gov identifier NCT04626635.ReferencesWaite JC, Wang B, Haber L, et al. Tumor-targeted CD28 bispecific antibodies enhance the antitumor efficacy of PD-1 immunotherapy. Sci Transl Med 2020;12:2325.Ethics ApprovalThis study was conducted in accordance with the principles of the Declaration of Helsinki and the International Conference on Harmonization Good Clinical Practice guidelines. The study protocol and all amendments were approved by the institutional review board/ethics committee at each participating study site.ConsentAll patients provided written informed consent.
- Published
- 2021
12. 923 Hedgehog signaling drives epithelial-to-mesenchymal transition, immune evasion, and anti-PD-1 resistance through coordinated upregulation of Wnt ligands and PGE2 synthesis
- Author
-
Michael Plebanek, Nicholas DeVito, Balamayooran Theivanthiran, Brent A. Hanks, Y-Van Nguyen, and Michael Sturdivant
- Subjects
Pharmacology ,Cancer Research ,Chemistry ,Immunology ,Anti pd 1 ,Wnt signaling pathway ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Evasion (ethics) ,Hedgehog signaling pathway ,Cell biology ,Immune system ,Oncology ,Downregulation and upregulation ,Molecular Medicine ,Immunology and Allergy ,Epithelial–mesenchymal transition ,RC254-282 - Abstract
BackgroundImmunotherapy resistance has been correlated with epithelial-to-mesenchymal transition (EMT),1 2 however our understanding of tumor-intrinsic mechanisms driving this immune evasive phenotype is lacking. We have previously shown that Wnt ligands are upregulated in anti-PD-1 resistant melanomas,3 and postulated that upstream transcriptional regulation of select EMT pathways may underpin these findings. The hedgehog signaling (HH) transcription factor Gli2 promotes EMT.MethodsGli2 was constitutively activated (Gli2CA ) in a BRAFV600EPTEN-/- murine cell line via an N-terminal truncating mutation and silenced using CRISPR-Cas9. Multi-parameter flow cytometry and RNAseq was utilized to evaluate the impact of Gli2 on the tumor immune microenvironment. Anti-PD-1 resistance studies were performed in Gli2CA and control tumors. Bioinformatics studies were conducted using the melanoma TCGA and Hugo et al databases.2ResultsWe found upregulation of Gli2 targets in patients with anti-PD-1-refractory metastatic melanoma as well as in an autochthonous BRAFV600EPTEN-/- melanoma model after escape from anti-PD-1. RNAseq and Western blot studies demonstrated Gli2CA to promote EMT and Wnt ligand production in addition to upregulated COX2 in BRAFV600EPTEN-/- melanoma. This finding was reversed by genetic ablation and pharmacologic inhibition of Gli2, implicating a previously undescribed role for Gli2 in modulating COX2. These data were consistent with a notable correlation between a Gli2 signature and a prostaglandin synthesis signature in human melanoma TCGA database. Flow cytometry analysis showed exclusion of cytolytic T and NK cells, a shift from cDC1s to cDC2s, and enhanced MDSC recruitment in Gli2CA tumors. Consistent with these findings, whole tumor RNAseq of Gli2CA tumors demonstrated a decrease in Cd3e, Prf1, and Xcr1 with a concomitant increase in Cxcl1, Cxcl2, Ccl2, Ptgs2, and Arg1 relative to control tumors. RNAseq of FACS-sorted DCs from Gli2CA tumors demonstrated a loss of cDC1-associated genes including Xcr1, Wdfy4, and Clec9a compared to DCs derived from control tumors. In-line with our previous results showing that Wnt5a promotes MDSC recruitment in a Yap-dependent manner,4 we found that Yap inhibition or Wnt5a deletion in the BRAFV600EPTEN-/-Gli2CA cell line diminished MDSC-recruiting chemokines. Further consistent with these findings, Gli2CA tumors resist anti-PD-1 antibody therapy.Abstract 923 Figure 1Gli2 in tumors promotes Wnt and prostaglandin signaling, generating an immunosuppressive microenvironmentConclusionsOur data demonstrates that the HH transcription factor Gli2 drives the development of a tolerogenic tumor microenvironment unfavorable to anti-PD-1 immunotherapy by coordinating the upregulation of Wnt ligand expression and prostaglandin synthesis (figure 1). We propose that HH gene signatures are worthy of further study as a guide for selecting Wnt ligand and prostaglandin inhibitors in future immunotherapy studies.AcknowledgementsThe authors would like to acknowledge the Duke Cancer Institute Flow Cytometry Core.ReferencesBagaev A, et al. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell 2021.Hugo W, et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 2016;165(1):35–44.DeVito NC, et al. Pharmacological Wnt ligand inhibition overcomes key tumor-mediated resistance pathways to anti-PD-1 immunotherapy. Cell Rep 2021;35(5):109071.Theivanthiran B, et al. A tumor-intrinsic PD-L1/NLRP3 inflammasome signaling pathway drives resistance to anti-PD-1 immunotherapy. J Clin Invest 2020;130(5):2570–2586.
- Published
- 2021
13. 794 Long-term anti-tumor preclinical efficacy of an optimized anti PD-1/IL-7 bifunctional antibody sustaining activation of progenitor stem-like CD8 TILs and disarming Treg suppressive activity
- Author
-
Caroline Mary, Isabelle Girault, Nicolas Poirier, Margaux Seite, Géraldine Teppaz, Aurore Morello, Justine Durand, Emmanuelle Wilhem, and Virginie Thepenier
- Subjects
Pharmacology ,Antitumor activity ,Cancer Research ,biology ,business.industry ,Immunology ,Anti pd 1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,chemistry.chemical_compound ,Oncology ,chemistry ,biology.protein ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Medicine ,Antibody ,business ,Bifunctional ,CD8 ,RC254-282 ,Progenitor - Abstract
BackgroundDespite the PD-(L)1 therapy success, a majority of patients remain resistant. PD-1+IL7R+ progenitors CD8 TILs is a key T-cell subset associated with durable PD-(L)1 therapy response. However, this subset may rapidly undergo apoptosis and/or being fully exhausted after PD-(L)1 blockade. Some cytokines have the potential to strengthen PD-(L)1 therapy by promoting T cell survival, however, their clinical developments are limited by a shortened half-life and systemic toxicity. To redirect immunotherapy to tumor-specific T cells, expressing PD1, we propose to selectively deliver the pro-survival IL-7 to PD-1+ T cells using a bifunctional anti-PD1/IL-7 mutein antibody. We previously described that the anti-PD1/IL-7v abrogated suppressive activity of human Treg. Here we evaluated its preclinical anti-tumor efficacy and how it promotes the response of PD1+IL7R+ tumor-specific T cells.MethodsProliferation, IFN-γ, IL-7R signaling, and NFAT assays were tested to determine the mechanism of this antibody. For the suppressive assay, CD4 Treg and autologous CD8 Teff were co-cultured. In vivo experiments were performed in hPD-1 KI immunocompetent or humanized immunodeficient mice.ResultsThe anti-PD1/IL-7v antibody design has been optimized with a monovalent approach to enhance its biological activity: (1) preserved PD-1 antagonist activity, (2) improved pSTAT5 IL7R signaling, and (3) enhanced in vivo drug exposure and antitumor efficacy. An IL7 mutein has been designed to improve activity on PD1+ T cells while sparing PD1neg T cells.Using a chronic antigen stimulation model, anti-PD1/IL-7v restores the proliferation & survival of both early and fully exhausted CD8+ or CD4+ T cells. Similarly, anti-PD1/IL-7v, but not anti-PD1 alone, reactivates exhausted TILs isolated from human resected tumors. Gene expression analysis by Nanostring showed increase cytotoxicity, antigen presentation, and chemokines signatures. In vivo, anti-PD1/IL-7v demonstrated high monotherapy efficacy (90%) in a PD-1 sensitive orthotopic immunocompetent mouse tumor model as well as in a PD-1 refractory tumor model with 70% of CR vs 15% for anti-PD-1 alone. A selective higher expansion of stem-like/progenitors CD8 TILs was observed after therapy with anti-PD1/IL-7v compared to anti-PD1. Memory immune response was demonstrated in 100% of cured mice after tumor rechallenge in the absence of new treatment in 3 different tumor models. Finally, using two different humanized mouse models implanted with human tumors (A549 or MDA-MB231), we confirmed significant preclinical monotherapy efficacy of the anti-PD-1/IL7v.ConclusionsThese data highlight the potential of anti-PD1/IL-7 bifunctional drug to overcome immunotherapy resistance and to promote durable anti-tumor efficacy by preferentially reinvigorating PD-1+IL7R+ stem-like progenitors CD8 T cells.
- Published
- 2021
14. Anti-Angiogenic Agent Combined with Anti-PD-1 Immunotherapy Showed Activity in Patients With Classical Hodgkin Lymphoma Who Have Failed Immunotherapy: A Retrospective Case Report Study
- Author
-
Shuna Yao, Jialin Ma, Junfeng Chu, Shuang Zhao, Yanyan Liu, Haiying Wang, Zheng Yan, and Zhihua Yao
- Subjects
camrelizumab ,Report study ,business.industry ,medicine.medical_treatment ,Anti pd 1 ,Anti angiogenic ,Immunology ,Case Report ,Immunotherapy ,RC581-607 ,anti-angiogenic agent ,PD-1 ,Cancer research ,medicine ,Classical Hodgkin lymphoma ,Immunology and Allergy ,In patient ,immunotherapy ,Immunologic diseases. Allergy ,business ,Hodgkin lymphoma ,apatinib - Abstract
BackgroundPD-1/PD-L1 inhibitor immunotherapy has showed impressive activity in various cancers, especially relapsed/refractory (r/r) classical Hodgkin lymphoma (cHL). However, acquired resistance is inevitable for most patients. Sometimes severe side effects also lead to treatment termination. When immunotherapy failed, alternative treatment options are limited. In the past few years, we have used the anti-angiogenic agent apatinib and PD-1 inhibitor camrelizumab to treat cHL patients who failed prior immunotherapy. In this study, we analyzed the data of these patients.Patients and MethodsPatients with r/r cHL who had failed immunotherapy and subsequently received apatinib-camrelizumab (AC) combination therapy were included in this study. Patient data were collected from medical records and follow-up system. The efficacy and safety of AC therapy were analyzed.ResultsSeven patients who failed immunotherapy were identified in our database, of which five patients acquired immunotherapy resistance and two patients experienced severe side effects. They received a combination of camrelizumab (200 mg every four weeks) and apatinib (425 mg or 250 mg per day). As of the cut-off date, these patients had received a median of 4 cycles (range, 2 - 31) of treatment. Two (2/7) patients achieved complete response, four (4/7) partial response, and one (1/7) stable disease. The median progression-free survival was 10.0 months (range, 2.0 – 27.8). Low-dose apatinib (250 mg) plus camrelizumab was well tolerated and had no unexpected side effects. Besides, no reactive cutaneous capillary endothelial proliferation was observed in AC-treated patients.ConclusionsLow dose apatinib plus camrelizumab might be a promising treatment option for r/r cHL patients who have failed immunotherapy. This combination treatment is worthy of further investigation in more patients including solid cancer patients who have failed immunotherapy.
- Published
- 2021
- Full Text
- View/download PDF
15. An evidence mapping and scientometric analysis of the top-100 most cited clinical trials of anti-PD-1/PD-L1 drugs to treat cancers
- Author
-
Jiarui Wu, Jinhui Tian, Shuzhen Shi, Ming Liu, Junhua Zhang, Yuan Yuan, and Ya Gao
- Subjects
medicine.medical_specialty ,Programmed Cell Death 1 Receptor ,RM1-950 ,Evidence mapping ,PD-1/PD-L1 ,B7-H1 Antigen ,Programmed cell death ligand 1 ,scientometric analysis ,PD-L1 ,Neoplasms ,medicine ,Humans ,cancer ,Statistical analysis ,Lung cancer ,Immune Checkpoint Inhibitors ,Pharmacology ,Clinical Trials as Topic ,clinical trials ,Evidence-Based Medicine ,biology ,business.industry ,Anti pd 1 ,Cancer ,General Medicine ,medicine.disease ,Clinical trial ,evidence mapping ,citations ,Treatment Outcome ,Bibliometrics ,Family medicine ,biology.protein ,Therapeutics. Pharmacology ,business - Abstract
Objective To gain a deeper understanding of the hot topics and future prospects of programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1) inhibitors treatment of cancer through scientometric analysis of the top-100 most cited clinical trials. Materials and methods We searched the Web of Science Core Collection database from 1980 to June 2019. Two reviewers independently screened the top-100 most cited clinical trials that defined by the National Institutes of Health starting from the most cited article. Title, year of publication, citations, type of cancer, and focused aspects of outcomes were extracted from included clinical trials. VOSviewer software (version 1.6.9) and Excel 2016 were used to do statistical analysis. The evidence mapping was used to present the relationship between cancers, drugs, citations, and outcomes, etc. RESULTS: The top-100 most cited clinical trials published from 2010 to 2018 in nine journals with high impact factor (IF) (IF2018:6.68-70.67), and Lancet Oncology (USA) published the most clinical trials (n = 29, IF2018 = 35.3856). The total number of citations of the top-100 most cited clinical trials was from 59 to 5606. 920 authors from 34 countries and 458 organizations participated in publishing the top-100 most cited clinical trials. The USA (n = 95) and Memorial Sloan-Kettering Cancer Center (n = 31) contributed the most publications. Based on the evidence mapping, there are 25 different types of cancers (e.g. lung cancer, melanoma, and renal cell cancer) and five focused aspects of outcomes (e.g. safety and efficacy). Conclusion The USA was the dominant country. Anti-PD-1/PD-L1 drugs were widely used to treat lung cancer, melanoma, renal cell cancer, and Hodgkin lymphoma. More exploration should be done to explore the use of anti-PD-1/PD-L1 drugs to treat more type of cancers in future research.
- Published
- 2021
16. Anti-Tumor Necrosis Factor Receptor 2 Antibody Combined With Anti-PD-L1 Therapy Exerts Robust Antitumor Effects in Breast Cancer
- Author
-
Wei Zhong, Yi Cheng, Qiang Fu, Qian Shen, Liu Huang, and Jin Tong
- Subjects
biology ,business.industry ,QH301-705.5 ,Anti pd 1 ,anti-PD-L1 therapy ,Cell Biology ,medicine.disease ,Cell and Developmental Biology ,TNFR2 ,Breast cancer ,breast cancer ,Cancer research ,biology.protein ,Medicine ,Anti tumor necrosis factor ,exert robust anti-tumor ,Antibody ,Biology (General) ,business ,Receptor ,TNFR2 antagonist ,Developmental Biology ,Original Research - Abstract
Breast cancer is a leading type of malignant tumor in women; however, the immunotherapy in breast cancer is still underappreciated. In this study, we demonstrated that tumor necrosis factor receptor 2 (TNFR2) is highly expressed in both breast tumor tissue and tumor-infiltrating immunosuppressive CD4+Foxp3+ regulatory T cells (Tregs). We found that TNFR2 antagonistic antibody reduced Foxp3 expression and the proliferation of Tregs and impaired the inhibitory effect of Tregs on CD4+CD25– effector T (Teff) cells in a dose-dependent manner. The treatment of anti-TNFR2 antibody not only inhibited the proliferation of breast tumor cells in vitro but also suppressed the tumorigenesis of murine mammary carcinoma 4T1 cells in vivo. Mice recovered from tumor growth also developed 4T1-specific immunity. Furthermore, we demonstrated that anti-TNFR2 antibody in combination with anti-PD-L1 exhibited augmented antitumor effects than monotherapy. Anti-TNFR2 treatment also tended to increase the expression of proinflammatory cytokines in tumor tissues. In conclusion, our study suggests that TNFR2 antagonist could potentially offer a clinical benefit as a single agent or in combination with immune checkpoint blockade treatment for breast cancer immunotherapy.
- Published
- 2021
- Full Text
- View/download PDF
17. 588 Caloric restriction sensitizes melanoma to anti-PD-1 immune checkpoint inhibition
- Author
-
Shuyang Qin, Alexander C. Chacon, Peter A. Prieto, Alexa Melucci, and Rachel Jewell
- Subjects
Pharmacology ,Cancer Research ,business.industry ,Melanoma ,Immunology ,Anti pd 1 ,Caloric theory ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Immune checkpoint ,Oncology ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business ,RC254-282 - Abstract
BackgroundIndividual response to immune checkpoint inhibition (ICI) in patients with metastatic melanoma varies from 10–40% for monotherapy and 50–60% with combination therapy [1]. Identification of adjuncts to ICI to further bridge treatment response to cure is imperative. We focus on caloric restriction (CR) as an adjunct to anti-PD-1 ICI given its inexpensive nature, relative ease of application, and increased tolerability as compared to fasting.Methods12-week C57BL/6J mice were inoculated with Yale University Mouse Melanoma (YUMM 1.7), randomized into diet groups and further randomized into αPD-1 and control (IgG) groups on day 12 (D12). Full diet mice ad lib fed while CR mice were 40% calorically restricted based on average daily food intake. Tumors were measured every 3 days with digital calipers (q3d). αPD-1 or control was intraperitoneally injected starting D12 continuing q3d for 7 injections. Mice were sacrificed and tumors harvested on D31. RNA sequencing was performed on CD45+ CD3+ T cells.ResultsUnder CR conditions, mice treated with αPD-1 had significantly smaller tumor volumes compared to the full diet cohort treated with αPD-1 (D22, 271.15 m3 vs. 336.72 m3, p=0.031) and persisted to harvest (D31, 600.96 m3 vs. 1039.84 m3, p=0.034). A significant difference in tumor volumes between CR αPD-1 and CR control treated cohorts was observed starting at D28 (439.34 m3 vs. 667.63 m3, p=0.005) and persisted to harvest (D31, 600.96 m3 vs. 884.08 m3, p=0.009). However, no significant difference in tumor growth under full diet conditions in murine cohorts treated with αPD-1 or control or separately between CR and full diet cohorts treated with control was observed.On pathway enrichment analysis inflammatory response, cytokine-mediated, response to interferon-gamma, and cell proliferation pathways were downregulated in the CR + αPD-1 cohort. Notable genes found in these pathways include B-cell linker protein (BLNK), tyrosine-protein kinase Lyn (LYN), SYK (spleen tyrosine kinase), toll-like receptor (TLR) TLR4, TLR7, and TLR8.ConclusionsCaloric restriction significantly sensitizes YUMM 1.7 murine melanoma to anti-PD-1 therapy resulting in decreased tumor growth. We show significant modulation of tumor growth in a murine tumor cell line, which has previously demonstrated limited response to αPD-1. In the present study, caloric restriction may decrease inflammation via downregulation of cytokine and toll-like receptor mediated pathways. Furthermore, caloric restriction may reverse the immunosuppressive tumor microenvironment and provide an inexpensive means to increase treatment response to anti-PD-1 therapy.ReferencesWard WH, Farma JM. Cutaneous Melanoma: Etiology and Therapy. Brisbane (AU): Codon Publications. 2017; Chapter 8.Ethics ApprovalThis study was approved by the University Committee on Animal Resources (UCAR), UCAR-2018-014.
- Published
- 2021
18. Immune checkpoint inhibitors (anti PD‐1 or anti PD‐L1) plus platinum‐etoposide versus platinum‐etoposide alone for first‐line treatment of extensive small cell lung cancer
- Author
-
Fanqi Wu, Ming Liu, Jinhui Tian, Fujian Song, Shuzhen Shi, Junhua Zhang, and Ya Gao
- Subjects
business.industry ,Immune checkpoint inhibitors ,Anti pd 1 ,chemistry.chemical_element ,First line treatment ,chemistry ,medicine ,Cancer research ,Pharmacology (medical) ,Non small cell ,business ,Platinum ,Etoposide ,medicine.drug - Abstract
Objectives: This is a protocol for a Cochrane Review (intervention). The objectives are as follows:. To evaluate the efficacy and safety of immune checkpoint inhibitors (anti PD-1 or anti PD-L1) plus platinum-etoposide compared with platinum-etoposide alone for the first-line treatment of extensive-stage small cell lung cancer (ES-SCLC).
- Published
- 2021
19. PIV and PILE Score at Baseline Predict Clinical Outcome of Anti-PD-1/PD-L1 Inhibitor Combined With Chemotherapy in Extensive-Stage Small Cell Lung Cancer Patients
- Author
-
Ran Zeng, Fang Liu, Chen Fang, Jin Yang, Lifeng Luo, Ping Yue, Beili Gao, Yuchao Dong, and Yi Xiang
- Subjects
Blood Platelets ,Male ,medicine.medical_specialty ,Lung Neoplasms ,Neutrophils ,medicine.medical_treatment ,Immunology ,PILE ,Gastroenterology ,Sensitivity and Specificity ,Severity of Illness Index ,Internal medicine ,medicine ,Immunology and Allergy ,Humans ,Lymphocyte Count ,Pan-Immune-Inflammation Value ,Immune Checkpoint Inhibitors ,Original Research ,Aged ,Randomized Controlled Trials as Topic ,Chemotherapy ,anti-PD-1/PD-L1 inhibitors ,business.industry ,Anti pd 1 ,RC581-607 ,Middle Aged ,Prognosis ,Small Cell Lung Carcinoma ,Treatment efficacy ,Progression-Free Survival ,Clinical trial ,Absolute neutrophil count ,Biomarker (medicine) ,biomarker ,Female ,small cell lung cancer ,Immunologic diseases. Allergy ,business ,PD-L1 inhibitor ,Extensive-stage small cell lung cancer - Abstract
ObjectivesThe objective of this study is to evaluate whether PIV (Pan-Immune-Inflammation Value) and PILE [a score derived from PIV, lactate dehydrogenase (LDH), and Eastern Cooperative Oncology Group Performance Status (ECOG PS)] can predict clinical outcome of anti-PD-1/PD-L1 inhibitor combined with chemotherapy in patients with extensive-stage (ES) small cell lung cancer (SCLC).MethodsA total of 53 patients with ES-SCLC in the control group of clinical trial (NCT03041311) were included in this study. PIV was calculated as follows: (neutrophil count × platelet count × monocyte count)/lymphocyte count. The PILE scores were composited based on PIV, LDH levels, and ECOG PS. The Kaplan–Meier method and Cox hazards regression models were used for survival analyses. Moreover, the predictive ability of PIV and PILE was validated in an independent real-world group consisting of 84 patients.ResultsPatients in the low PIV group (PIV < median) had longer progression-free survival (PFS) and overall survival (OS) than those in the high PIV group (PIV ≥ median), along with the HR, which was 2.157 and 2.359, respectively (PFS HR 95% CI: 1.181–3.940, p = 0.012; OS HR 95% CI: 1.168–4.762, p = 0.020). High PILE score was observed relating to worse treatment efficacy (disease control rate (DCR): 84.21% vs. 100%, p = 0.047; durable clinical benefit (DCB) rate: 10% vs. 48.5%, p = 0.060) and poor clinical outcome (median PFS: 4.75 vs. 5.53 m, p = 0.043; median OS: 7.13 vs. 15.93 m, p = 0.002). Similar results were obtained about the predictive and prognostic abilities of PIV and PILE scores in the validation group.ConclusionsHigh PIV and high PILE were correlated with worse clinical outcomes in ES-SCLC patients treated with anti-PD-1/PD-L1 inhibitor combined with chemotherapy, reflecting that PIV and PILE might be useful to identify patients unlikely to benefit from anti-PD-1/PD-L1 therapy.
- Published
- 2021
20. Advanced hepatocellular carcinoma treated by radiofrequency ablation combined with oncolytic virus and anti-PD-1 antibody therapy: a case report and literature review
- Author
-
Ximing Sun, Feng Xia, Zongtao Song, Jun Pei, and Anqing Xie
- Subjects
Medicine (General) ,anti-PD-1 antibody ,Carcinoma, Hepatocellular ,Radiofrequency ablation ,medicine.medical_treatment ,Case Reports ,Biochemistry ,law.invention ,R5-920 ,law ,recombinant human adenovirus type 5 ,medicine ,Humans ,Complete response ,oncolytic virus ,Radiofrequency Ablation ,biology ,business.industry ,Biochemistry (medical) ,Anti pd 1 ,Liver Neoplasms ,Patient survival ,Cell Biology ,General Medicine ,Immunotherapy ,medicine.disease ,digestive system diseases ,Oncolytic virus ,Oncolytic Viruses ,Hepatocellular carcinoma ,biology.protein ,Cancer research ,advanced hepatocellular carcinoma ,Antibody ,business - Abstract
The development of an effective therapy for advanced hepatocellular carcinoma (HCC) represents an important global concern. In recent years, the combination of multiple treatment methods with immunotherapy has achieved great progress in patients with advanced HCC. Patient survival has been significantly prolonged, but cases of complete response (CR) remain rare. Here, we report two cases in which CR was achieved by radiofrequency ablation combined with an oncolytic virus (recombinant human adenovirus type 5) and anti-programmed cell death protein 1 antibody. Additionally, a literature review is presented to describe similar advancements in this field and explore viable methods for the treatment of advanced HCC.
- Published
- 2021
21. Predictive Role of Prior Radiotherapy and Immunotherapy-Related Adverse Effects in Advanced NSCLC Patients Receiving Anti-PD-1/L1 Therapy
- Author
-
Soo Han Kim, Seung Joon Kim, Sung Kyoung Kim, Jin Woo Kim, Jeong Uk Lim, Hye Seon Kang, Ju Sang Kim, Chang Dong Yeo, and Chang-Min Choi
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Prior Radiotherapy ,medicine.medical_treatment ,Immune checkpoint inhibitors ,Anti pd 1 ,adverse event ,General Medicine ,Immunotherapy ,medicine.disease ,Article ,Radiation therapy ,lung cancer ,Internal medicine ,medicine ,Medicine ,In patient ,immunotherapy ,Adverse effect ,business ,Lung cancer ,radiotherapy - Abstract
The present study evaluated the impact of prior radiotherapy (RT) on patients with advanced non-small cell lung cancer (NSCLC) receiving therapy with immune checkpoint inhibitors (ICIs) and further assessed the prognostic factors in patients receiving both RT and ICI. Patients diagnosed with NSCLC at the Catholic Medical Center and Asan Medical Center between January 2016 and October 2020 and who received immunotherapy were retrospectively reviewed. Among 240 patients, poor Eastern Cooperative Oncology Group (ECOG) score, high PD-L1 expression, and ICI-related adverse events (AE) were significantly associated with progression-free survival (PFS) (HR, 2.654, 95% CI, 1.484–4.749, p = 0.001, HR, 0.645, 95% CI, 0.449–0.926, p = 0.017, HR, 0.430, 95% CI, 0.229–0.808, p = 0.009, respectively). Among patients who received both RT and immunotherapy, poor ECOG status, squamous cell carcinoma, and ICI-related AE were significant factors associated with poor PFS (HR, 2.430, 95% CI, 1.464–4.034, HR, 0.667, 95% CI, 0.455–0.978, p = 0.038, HR, 0.520, 95% CI, 0.284–0.953, p = 0.034, respectively). The present study showed that prior RT showed no significant independent association with primary outcomes in patients with advanced NSCLC receiving immunotherapy. In patients who received both RT and immunotherapy, clinical parameters, including ICI-related AEs, were independently predictive of PFS.
- Published
- 2021
22. Phase I/Ib Clinical Trial of Sabatolimab, an Anti-TIM-3 Antibody, Alone and in Combination with Spartalizumab, an Anti-PD-1 Antibody, in Advanced Solid Tumors
- Author
-
Sofie Wilgenhof, Armando Santoro, Luigi Manenti, Nicolas Mach, Chia-Chi Lin, A. Xyrafas, Tyler Longmire, Patrick M. Forde, Haiying Sun, Philippe L. Bedard, Toshihiko Doi, Catherine Anne Sabatos-Peyton, Aung Naing, Hans Gelderblom, Giuseppe Curigliano, David Tai, F. Stephen Hodi, John Sarantopoulos, and Sabine Gutzwiller
- Subjects
Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Colorectal cancer ,Programmed Cell Death 1 Receptor ,Antibodies, Monoclonal, Humanized ,Gastroenterology ,Young Adult ,Neoplasms ,Internal medicine ,medicine ,Humans ,In patient ,Adverse effect ,Hepatitis A Virus Cellular Receptor 2 ,Immune Checkpoint Inhibitors ,Aged ,Neoplasm Staging ,Aged, 80 and over ,ddc:616 ,biology ,business.industry ,Melanoma ,Mucin ,Anti pd 1 ,Antibodies, Monoclonal ,Middle Aged ,medicine.disease ,Clinical trial ,Drug Combinations ,Oncology ,biology.protein ,Female ,Antibody ,business - Abstract
Purpose:Sabatolimab (MBG453) and spartalizumab are mAbs that bind T-cell immunoglobulin domain and mucin domain-3 (TIM-3) and programmed death-1 (PD-1), respectively. This phase I/II study evaluated the safety and efficacy of sabatolimab, with or without spartalizumab, in patients with advanced solid tumors.Patients and Methods:Primary objectives of the phase I/Ib part were to characterize the safety and estimate recommended phase II dose (RP2D) for future studies. Dose escalation was guided by a Bayesian (hierarchical) logistic regression model. Sabatolimab was administered intravenously, 20 to 1,200 mg, every 2 or 4 weeks (Q2W or Q4W). Spartalizumab was administered intravenously, 80 to 400 mg, Q2W or Q4W.Results:Enrolled patients (n = 219) had a range of cancers, most commonly ovarian (17%) and colorectal cancer (7%); patients received sabatolimab (n = 133) or sabatolimab plus spartalizumab (n = 86). The MTD was not reached. The most common adverse event suspected to be treatment-related was fatigue (9%, sabatolimab; 15%, combination). No responses were seen with sabatolimab. Five patients receiving combination treatment had partial responses (6%; lasting 12–27 months) in colorectal cancer (n = 2), non–small cell lung cancer (NSCLC), malignant perianal melanoma, and SCLC. Of the five, two patients had elevated expression of immune markers in baseline biopsies; another three had >10% TIM-3–positive staining, including one patient with NSCLC who received prior PD-1 therapy.Conclusions:Sabatolimab plus spartalizumab was well tolerated and showed preliminary signs of antitumor activity. The RP2D for sabatolimab was selected as 800 mg Q4W (alternatively Q3W or Q2W schedules, based on modeling), with or without 400 mg spartalizumab Q4W.
- Published
- 2021
23. Clinical Activity and Safety of Penpulimab (Anti-PD-1) With Anlotinib as First-Line Therapy for Unresectable Hepatocellular Carcinoma: An Open-Label, Multicenter, Phase Ib/II Trial (AK105-203)
- Author
-
Shunchang Jiao, Liangfang Shen, Yuxian Bai, Aimin Zang, Chunhong Hu, Li Bai, Chunmei Bai, Chun Han, Qun Qin, Shizhong Yang, and Sisi Ye
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,ECOG Performance Status ,Gastroenterology ,immune checkpoint inhibitors ,03 medical and health sciences ,0302 clinical medicine ,First line therapy ,Internal medicine ,Clinical endpoint ,medicine ,anlotinib ,Adverse effect ,RC254-282 ,Original Research ,business.industry ,Anti pd 1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,hepatocellular carcinoma ,medicine.disease ,BCLC Stage ,antiangiogenics ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Open label ,business ,first-line treatment ,penpulimab - Abstract
ObjectiveThis study aims to assess the efficacy and safety of penpulimab (a humanized anti-PD-1 IgG1 antibody) with anlotinib in the first-line treatment of Chinese patients with uHCC.MethodsIn this open-label multicenter phase Ib/II trial, patients with histologically or cytologically confirmed uHCC, without previous systemic treatment, aged 18–75 years old, classified as BCLC stage B (not amenable for locoregional therapy) or C, with Child–Pugh score ≤7 and ECOG performance status ≤1 were enrolled. Patients received penpulimab [200 mg intravenous (i.v.) Q3W] and oral anlotinib (8 mg/day, 2 weeks on/1 week off). The primary endpoint was objective response rate (ORR). Secondary endpoints included safety, disease control rate (DCR), progression-free survival (PFS), time to progression (TTP), duration of response (DoR), and overall survival (OS). This trial is registered with ClinicalTrials.gov (NCT04172571).ResultsAt the data cutoff (December 30, 2020), 31 eligible patients had been enrolled and treated with a median follow-up of 14.7 months (range, 1.4–22.1). The ORR was 31.0% (95% CI, 15.3–50.8%), and the DCR was 82.8% (95% CI, 64.2–94.2%). The median PFS and TTP for 31 patients were 8.8 months (95% CI, 4.0–12.3) and 8.8 months (95% CI, 4.0–12.9) respectively. The median OS was not reached; the 12-month OS rate was 69.0% (95% CI, 48.9–82.5%). Only 19.4% (6/31) of patients had grade 3/4 treatment-related adverse events (TRAEs).ConclusionPenpulimab plus anlotinib showed promising anti-tumor activity and a favorable safety profile as first-line treatment of patients with uHCC.
- Published
- 2021
- Full Text
- View/download PDF
24. Prognostic Impact of Sarcopenia and Radiotherapy in Patients With Advanced Gastric Cancer Treated With Anti-PD-1 Antibody
- Author
-
Nalee Kim, Seung Tae Kim, Jeeyun Lee, Woo Kyoung Jeong, Kyoung-Mee Kim, Jeong Il Yu, Won Ki Kang, Do Hoon Lim, and Jung Yong Hong
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Sarcopenia ,Neutrophils ,medicine.medical_treatment ,Immunology ,Programmed Cell Death 1 Receptor ,Gastroenterology ,radiation therapy ,Antibodies ,03 medical and health sciences ,0302 clinical medicine ,Stomach Neoplasms ,Internal medicine ,medicine ,Immunology and Allergy ,Humans ,In patient ,Lymphocytes ,Immune Checkpoint Inhibitors ,Original Research ,Aged ,business.industry ,gastric cancer ,Anti pd 1 ,Significant difference ,Advanced gastric cancer ,RC581-607 ,Middle Aged ,medicine.disease ,Prognosis ,Predictive value ,Radiation therapy ,030104 developmental biology ,inflammation ,030220 oncology & carcinogenesis ,Cohort ,Female ,Microsatellite Instability ,immunotherapy ,Immunologic diseases. Allergy ,business - Abstract
BackgroundWe explored the combined effects of sarcopenia (SAR) and radiotherapy (RT) on outcomes in patients with advanced gastric cancer (AGC) treated with immune-checkpoint blockade (ICB).MethodsAmong 185 patients with AGC treated with ICB, we defined SAR as skeletal muscle index ResultsWe frequently observed hNLR in patients with SAR (53% vs. 35%, p = 0.02). The median overall survival (OS) for the entire cohort was 5 months. Stratification by risk factors of SAR or hNLR revealed a significant difference in median OS (0 [N = 60] vs. 1 [N = 76] vs. 2 [N = 49]: 7.6 vs. 6.4 vs. 2.2 months, p < 0.001). Patients with microsatellite instability-high (MSI-H, N = 19) or Epstein-Barr virus (EBV)-positive tumors (N = 13) showed favorable outcomes compared to those with microsatellite stable (MSS, N = 142) tumors (median OS, not reached vs. 16.8 vs. 3.8 months, respectively). The benefit of RT was evident in patients with both SAR and hNLR (median OS, 3.1 vs. 1.3 months, p = 0.02) and MSS/EBV-negative tumor (median OS, 6.5 vs. 3.5 months, p = 0.03), but outcomes after RT in MSI-H tumor were not significantly different. In multivariable analysis, SAR/hNLR, molecular subtypes, and a history of RT were associated with OS (all p < 0.05).ConclusionsWe demonstrated the negative predictive value of SAR/hNLR on outcomes after ICB for AGC, and the history of RT could overcome the negative impact of SAR/hNLR and the MSS/EBV-negative subtype.
- Published
- 2021
25. Lactate dehydrogenase and baseline markers associated with clinical outcomes of advanced esophageal squamous cell carcinoma patients treated with camrelizumab (SHR‐1210), a novel anti‐PD‐1 antibody
- Author
-
Hongnan Mo, Qun Li, Xi Wang, Bo Lan, Xuelian Chen, Dawei Wu, Binghe Xu, Bo Zhang, and Jing Huang
- Subjects
0301 basic medicine ,Male ,programmed cell death‐1 ,Cancer Research ,Esophageal Neoplasms ,Immune checkpoint inhibitors ,medicine.medical_treatment ,immune checkpoint inhibitor ,markers ,Gastroenterology ,Esophageal squamous cell carcinoma ,chemistry.chemical_compound ,Leukocyte Count ,0302 clinical medicine ,Medicine ,biology ,Hazard ratio ,General Medicine ,Middle Aged ,Prognosis ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,C-Reactive Protein ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Toxicity ,Cohort ,Disease Progression ,Administration, Intravenous ,Female ,Original Article ,Antibody ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Antibodies, Monoclonal, Humanized ,lcsh:RC254-282 ,Drug Administration Schedule ,03 medical and health sciences ,Internal medicine ,Lactate dehydrogenase ,Humans ,Aged ,Retrospective Studies ,L-Lactate Dehydrogenase ,business.industry ,Anti pd 1 ,lactate dehydrogenase ,Immunotherapy ,Original Articles ,Survival Analysis ,Peripheral blood ,030104 developmental biology ,chemistry ,Cancer research ,biology.protein ,business ,Biomarkers - Abstract
e15559 Background: A small proportion of patients with advanced esophageal squamous cell carcinoma (ESCC) could benefit from immune checkpoint inhibitors, and reliable peripheral blood biomarkers for outcomes of anti-PD-1 immunotherapy were not identified in ESCC. Methods: A total of 43 patients were retrospectively reviewed in the ESCC cohort of a phase I trial from our center. All patients received intravenous camrelizumab (SHR-1210), a novel anti-PD-1 antibody, at a dose of 60 mg, 200 mg or 400 mg (4-week interval after first dose followed by a 2-week schedule) and repeated every two weeks until disease progression or intolerable toxicity. The associations between lactate dehydrogenase (LDH) as well as other peripheral blood biomarkers at baseline and the efficacy of camrelizumab were also investigated. Results: With a median follow-up of 19.6 months, the overall response rate was 25.6% (11/43), including one complete response. Median progression-free survival (PFS) and overall survival were 2.0 months (95% CI: 0-4.1 months) and 8.0 months (95% CI: 7.2-8.8 months), respectively. Notably, four patients achieved a PFS exceeding 12 months, including three patients with a long-lasting duration of response over 1 year. Patients with an elevated baseline lactate dehydrogenase had lower tumor response rates (8.3% vs. 32.3%, p = 0.02) as well as shorter PFS (median: 1.8 vs. 4.0 months; HR 0.39, p = 0.002) and overall survival (median: 4.2 vs. 10.4 months; HR 0.22, p < 0.0001) compared with patients with normal levels. An increase of lactate dehydrogenase level during treatment was significantly associated with disease progression (p = 0.014). Multivariate Cox analysis identified LDH (HR = 0.18), C-reactive protein (HR = 0.27), number of involved organs (HR = 0.31), absolute monocyte count (HR = 0.33) and Eastern Cooperative Oncology Group performance status (HR = 0.36) as independent prognostic factors. Conclusions: Serum LDH, as is readily available in routine clinical practice, is a potential marker for response and a powerful independent factor for survival in advanced ESCC patients receiving anti-PD-1 treatment.
- Published
- 2019
26. A Patient with Nivolumab-related Fulminant Type 1 Diabetes Mellitus whose Serum C-peptide Level Was Preserved at the Initial Detection of Hyperglycemia
- Author
-
Jun Saitou, Tomoko Takiguchi, Haremaru Kubo, Rei Hirose, Sho Katsuragawa, Hiroko Yukawa, Naoko Yamamoto, Hitoshi Arioka, Tetsuo Nishikawa, Yoshitomo Hoshino, Takashi Sunouchi, Yuya Tsurutani, and Masahiro Ichikawa
- Subjects
Blood Glucose ,Male ,medicine.medical_specialty ,anti-PD-1 antibody ,Fulminant ,medicine.medical_treatment ,Case Report ,fulminant type 1 diabetes mellitus ,030204 cardiovascular system & hematology ,Gastroenterology ,Plasma glucose level ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Renal cell carcinoma ,Internal medicine ,Internal Medicine ,Medicine ,Humans ,Insulin ,Carcinoma, Renal Cell ,Aged ,nivolumab ,Type 1 diabetes ,C-Peptide ,business.industry ,C-peptide ,Anti pd 1 ,Antibodies, Monoclonal ,General Medicine ,medicine.disease ,Kidney Neoplasms ,Diabetes Mellitus, Type 1 ,chemistry ,Hyperglycemia ,030211 gastroenterology & hepatology ,Nivolumab ,business - Abstract
A 77-year-old-man with renal cell carcinoma who was undergoing nivolumab treatment visited our department due to hyperglycemia; his plasma glucose level was 379 mg/dL. Although his serum C-peptide immunoreactivity (CPR) level was preserved (5.92 ng/mL), we suspected an onset of fulminant type 1 diabetes mellitus (FT1DM) and immediately started insulin therapy. His CPR levels gradually decreased and were depleted within 1 week. We later discovered that the patient's casual CPR level had been abnormally high (11.78 ng/mL) 2 weeks before his admission. Hence, the possibility of FT1DM in hyperglycemic patients undergoing nivolumab treatment should not be excluded, even with a preserved CPR level.
- Published
- 2019
27. Reactive capillary hemangiomas: a novel dermatologic toxicity following anti-PD-1 treatment with SHR-1210
- Author
-
Lanying Ma, Hongtu Zhang, Hongnan Mo, Jing Huang, Bo Lan, Xuelian Chen, Dong Qu, Binghe Xu, Dawei Wu, and Xi Wang
- Subjects
Cancer Research ,medicine.medical_specialty ,SHR-1210 ,Context (language use) ,Gastroenterology ,lcsh:RC254-282 ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Adverse effect ,business.industry ,Capillary hemangioma ,Anti pd 1 ,Clinical course ,anti-tumor efficacy ,Reactive capillary hemangiomas ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Dermatologic toxicity ,skin toxicity ,Phase i study ,Oncology ,030220 oncology & carcinogenesis ,Cohort ,Original Article ,business - Abstract
Objective SHR-1210 is a new and promising anti-PD-1 agent for solid tumors. During the phase I study of SHR-1210, we encountered a novel but prevalent immune-related dermatologic toxicity: reactive capillary hemangiomas (RCHs). Thus we tried to summarize the features of RCHs and estimate their relationship with tumor response. Methods This prospective observational study systematically enrolled 98 patients with advanced solid tumors from April 27th, 2016 to June 8th, 2017 in the context of the phase I clinical study of SHR-1210. This report focused on the skin toxicities. Patients underwent entire skin inspection every two weeks while taking medication. The clinical course of RCHs was recorded and their association with tumor response was estimated. The data cut-off date was November 15th, 2017.Results After a median follow-up of 242 (range, 29–567) days, RCHs were observed in 85.7% (84/98) of patients on cutaneous/mucosal surfaces; 84.5% (71/84) of the RCHs were evaluated as grade 1 adverse events. No grade 3 or 4 RCHs were observed. The time of onset of RCHs was dose dependent and shortest in the 400 mg-dose cohort (P < 0.001). Spontaneous and complete regression of RCHs was observed both during and after treatment. The objective response rate of tumors for patients with RCHs was 28.9% (24/83). However, no responders were observed among the patients without RCHs.Conclusions RCHs were prevalent but manageable during treatment with SHR-1210. It might add to the expanding literature regarding immune-related dermatologic adverse events.
- Published
- 2019
28. MULTITARGET ANTI-PD-1/PD-L1/CD19/CD25/CD38 THERAPY USING CHELATE NANOCOMPLEX MSC-428 IN ONCOPATHOLOGY
- Author
-
E. S. Buryachkovsky, A. V. Artemov, and O. V. Lukyanchuk
- Subjects
biology ,oncopathology ,Chemistry ,Anti pd 1 ,lcsh:R ,multitarget therapy ,lcsh:Medicine ,chemical and pharmacologic phenomena ,CD38 ,Combinatorial chemistry ,CD19 ,lcsh:Biology (General) ,PD-L1 ,biology.protein ,Chelation ,nanocomplexes ,IL-2 receptor ,lcsh:QH301-705.5 - Abstract
Over the past decade, there has been a deepening of ideas about the relationship between the tumor and the hosts immune system, as evidenced by the 2018 Nobel Prize in medicine and physiology for studying the role of the so-called. Immune checkpoints in neoplasm care from an attack of the hosts immune system. This mechanism is associated with the appearance on the surface of tumor cells of specific proteins PD-1/PD-L-1, which prevent contact with the killer lymphocyte receptor. Chelate nanocomplexes (MSC-428) target PD-1/PD-L-1, i. e. affect the immune checkpoints directly. MSC-428 molecules attack 4 target proteins: PD-1/PD-L1, CD19, CD25, CD38, which leads to the normalization of surface antigens CD3, CD4, CD8, CD16, CD45, CD95 lymphocytes, phagocytosis systems and immunoglobulins of class A, M, G. The proposed strategy of treatment against PD-1/PD-L-1 with the use of metal-containing nanocomplex MSC428 expands the possibilities of influencing immune checkpoints. Through conformational change in the 3-dimensional structure of the receptor proteins PD-1, the drug prevents them from binding to PD-L1 in tumor cells. This increases the expression of CD25 on T-lymphocytes, normalizes the production of IL-2, which increases the activity of T-killers against tumor cells. The drug reduces the activity of CD19 molecules, which shield tumor antigens and prevent the attack of cytotoxic T cells. As a result of inhibition of ZAP-70 kinase, CD38 expression is reduced, which increases the bodys antitumor response and mobilizes the cytotoxic potential of CD8 and CD16 T lymphocytes, preventing the development of tumor cell resistance. The restoration of their activity largely predetermines the normalization of other lymphocyte subpopulations, in particular, expressing CD3, CD4, CD8, CD16, CD95, as well as the restoration of the phagocytosis system, production of immunoglobulins of class A, M, G. Such a complex effect ensures the construction of a coherent and effective strategy for immunotherapy of patients with oncopathology.
- Published
- 2018
29. Phase I study of pucotenlimab (HX008), an anti-PD-1 antibody, for patients with advanced solid tumors
- Author
-
Yanchun Meng, Xichun Hu, Wenhua Li, Jian Zhang, Shuiping Gao, and Rujiao Liu
- Subjects
0301 basic medicine ,safety ,medicine.drug_class ,Monoclonal antibody ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Pharmacokinetics ,Immunoglobulin g4 ,Programmed cell death 1 ,Medicine ,RC254-282 ,Original Research ,Mutation ,biology ,business.industry ,Anti pd 1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Phase i study ,Fc domain ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,pucotenlimab ,anti-PD-1 ,solid tumor ,business ,pharmacokinetics - Abstract
Background: Pucotenlimab is a humanized immunoglobulin G4 (IgG4) anti programmed cell death protein 1 (anti-PD-1) monoclonal antibody (mAb) with a S228P hinge mutation and an engineered Fc domain. Preclinical data suggests that pucotenlimab exerts antitumor effects. In this phase I study, which was prospectively registered on www.chinadrugtrials.org.cn (CTR20180125), the safety, maximum tolerated dose, preliminary antitumor activity, pharmacokinetics, and immunogenicity of pucotenlimab were evaluated in patients with advanced solid tumors. Methods: Patients with advanced solid tumors refractory to standard therapies were recruited. In a 3+3 dose escalation study, 13 patients received pucotenlimab intravenously every 3 weeks (Q3W) until disease progression or unacceptable toxicity occurred at doses of 1 mg/kg, 3 mg/kg, 10 mg/kg, and 200 mg. 17 additional patients were assigned in the expansion period. Results: A total of 30 patients were enrolled. No dose-limiting toxicity was observed. The maximum tolerated dose was not reached. The most common treatment-related adverse events of any grade were proteinuria (40%), fatigue (36.7%), weight loss (26.7%), fever (26.7%), increased aspartate aminotransferase (26.7%), rash (23.3%), and anorexia (20.0%). Partial responses occurred in five patients, with an objective response rate of 16.7%. Pharmacokinetics analysis showed rapid absorption followed by slow terminal elimination, with a mean half-life of 17.1–23.5 days across all dose groups. Conclusions: Pucotenlimab had an acceptable toxicity profile at doses up to 10 mg/kg and the maximum tolerated dose was not reached. Based on the pharmacokinetics, efficacy, and safety profile, 3 mg/kg Q3W or 200 mg Q3W are optimal for further drug development.
- Published
- 2021
30. Anti-PD-1 Immunotherapy Improves the Efficacy of Hepatic Artery Infusion Chemotherapy in Advanced Hepatocellular Carcinoma
- Author
-
Liang-He Lu, Qi-Jiong Li, Lie Zheng, Wei Wei, Minshan Chen, Wenping Lin, Ming Shi, Shao Hua Li, Xuqi Sun, Jie Mei, and Rong Ping Guo
- Subjects
medicine.medical_specialty ,Treatment response ,Combination therapy ,medicine.medical_treatment ,Gastroenterology ,hepatic artery infusion chemotherapy ,combination therapy ,03 medical and health sciences ,0302 clinical medicine ,FOLFOX ,Internal medicine ,medicine ,programmed cell death protein-1 ,Journal of Hepatocellular Carcinoma ,Original Research ,Chemotherapy ,business.industry ,Anti pd 1 ,hepatocellular carcinoma ,Immunotherapy ,medicine.disease ,Artery infusion ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,030211 gastroenterology & hepatology ,business ,medicine.drug - Abstract
Jie Mei,1,2,* Shao-Hua Li,1,2,* Qi-Jiong Li,1,2,* Xu-Qi Sun,1,2 Liang-He Lu,1,2 Wen-Ping Lin,1,2 Lie Zheng,1,3 Min-Shan Chen,1,2 Ming Shi,1,2 Wei Wei,1,2 Rong-Ping Guo1,2 1Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, People’s Republic of China; 2Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People’s Republic of China; 3Department of Medical Imaging, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People’s Republic of China*These authors contributed equally to this workCorrespondence: Rong-Ping Guo; Wei Wei Email guorp@sysucc.org.cn; weiwei@sysucc.org.cnBackground: Hepatic artery infusion chemotherapy (HAIC) and anti-programmed cell death protein-1 (PD-1) immunotherapy have shown promising outcomes in patients with advanced hepatocellular carcinoma (HCC), respectively. However, the combination of the two treatments has not been reported. In this study, we compared the efficacy of HAIC combined with anti-PD-1 immunotherapy (HAICAP) and HAIC in patients with advanced HCC.Methods: Between November 2018 and December 2019, advanced HCC patients that were treated with either HAICAP or HAIC were retrospectively recruited and reviewed for eligibility. Efficacy was evaluated according to tumor response and survival.Results: As a result, 229 patients were included in this study. Patients were divided into HAICAP group (n = 81) and HAIC group (n = 148) accordingly. The follow-up time ranged from 1.0 to 21.6 months, with a median of 11.0 months. The median overall survival was 18.0 months in the HAICAP group and 14.6 months in the HAIC group (p = 0.018; HR = 0.62; 95% CI 0.34– 0.91). The median progression-free survival was 10.0 months in the HAICAP group and 5.6 months in the HAIC group (p = 0.006; HR = 0.65; 95% CI 0.43– 0.87). The disease control rate in overall response (83% vs 66%; p = 0.006) and intrahepatic response (85% vs 74%, respectively; p = 0.045) were higher in the HAICAP group than in the HAIC group.Conclusion: In comparison to HAIC, HAICAP was associated with a better treatment response and survival benefits for patients with advanced HCC.Keywords: hepatocellular carcinoma, hepatic artery infusion chemotherapy, programmed cell death protein-1, FOLFOX, combination therapy
- Published
- 2021
31. Real-World Outcomes and Clinical Predictors of Immune Checkpoint Inhibitor Monotherapy in Advanced Lung Cancer
- Author
-
Manoj Kumar, Amit Kulkarni, Manish R. Patel, Beibei Xu, Daniel F. Pease, Shilvi P Joshi, Shijia Zhang, and Ryan Shanley
- Subjects
0301 basic medicine ,anti-PD-L1 ,Immune checkpoint inhibitors ,medicine.medical_treatment ,non-small cell lung cancer (NSCLC) ,small-cell lung cancer (SCLC) ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Lung cancer ,RC254-282 ,Non-small-cell lung cancer (NSCLC) ,business.industry ,Anti pd 1 ,Real world outcomes ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunotherapy ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,anti-PD-1 ,Original Article ,immunotherapy ,business - Abstract
Background: Immune checkpoint inhibitors (ICIs) have changed the treatment paradigm of advanced-stage non-small-cell lung cancer (NSCLC) and small-cell lung cancer (SCLC). The aim of this study was to evaluate the effectiveness and tolerance of ICIs in a real-world patient population and to investigate the predictive factors associated with survival outcomes. Methods: Medical records of patients with advanced lung cancer who started ICI monotherapy were reviewed for data collection. Treatment outcomes included objective response rate, progression-free survival (PFS), and overall survival (OS). Immune-related adverse events (irAEs) were assessed. Multiple Cox regression models were fit to investigate the predictive factors for survival outcomes. Results: We included 220 patients (median 66.5 years). Seventy-nine (35.9%) patients had Eastern Cooperative Oncology Group (ECOG) performance-status (PS) score ⩾2. Median follow-up was 11.4 months. In NSCLC, median PFS was 3.8 months (4.7 months for first line and 3.7 months for subsequent line). Median OS was 12.4 months (15.6 months for first line therapy and 11.5 months for subsequent line). In SCLC, median PFS was 1.8 months, and median OS was 4.6 months. A quarter of patients developed irAEs. There was 1 disease flare among 17 patients with pre-existing autoimmune diseases. ECOG PS of 0 to 1 and body mass index (BMI) ⩾ 25 kg/m2 (but not occurrence of irAE) were independently associated with improved OS in NSCLC, with a hazard ratio of 0.41 (95% confidence interval [CI], 0.29-0.59) and 0.62 (95% CI, 0.44-0.87), respectively. Conclusions: The clinical benefit of ICIs appears to persist in a real-world population of relatively older age, including those with poor PS and pre-existing autoimmune diseases. ECOG PS of 0 to 1 and BMI ⩾ 25 kg/m2 were independently associated with improved OS.
- Published
- 2021
32. Anti-PD-1 plus anti-VEGF therapy in multiple intracranial metastases of a hypermutated, IDH wild-type glioblastoma
- Author
-
Wenbin Ma, Xiaopeng Guo, Yu Wang, and Shishuai Wang
- Subjects
Anti vegf ,Cancer Research ,Bevacizumab ,business.industry ,Anti pd 1 ,Wild type ,medicine.disease ,Text mining ,Oncology ,Cancer research ,Medicine ,Neurology (clinical) ,business ,Letters to the Editor ,medicine.drug ,Glioblastoma - Published
- 2021
33. Anti-PD-1 antibody HX008 combined with oxaliplatin plus capecitabine for advanced gastric or esophagogastric junction cancer: a multicenter, single-arm, open-label, phase Ib trial
- Author
-
Jianming Xu, Yuxian Bai, Chenyu Mao, Yiwei Dou, Nong Xu, Hong Sui, Qian Jiang, Xiaofei Wang, and Rongrui Liu
- Subjects
0301 basic medicine ,China ,medicine.drug_class ,Immunology ,Monoclonal antibody ,Capecitabine ,03 medical and health sciences ,0302 clinical medicine ,Stomach Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Immunology and Allergy ,Medicine ,Esophagogastric junction ,RC254-282 ,Original Research ,hx008 ,business.industry ,capecitabine ,gastric cancer ,Anti pd 1 ,oxaliplatin ,pd-1 ,Antibodies, Monoclonal ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC581-607 ,medicine.disease ,Oxaliplatin ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Monoclonal ,Cancer research ,Esophagogastric Junction ,Open label ,Immunologic diseases. Allergy ,business ,Research Article ,medicine.drug - Abstract
Anti-PD-1 monoclonal antibody is approved as an option for third-line treatment of advanced gastric and gastroesophageal junction (G/GEJ) cancer in several countries, but no anti-PD-1 monoclonal antibody treatment is yet approved for first-line treatment of advanced G/GEJ cancer. We report a phase Ib trial of HX008, a highly selective, humanized anti-programmed death-1 monoclonal antibody, plus oxaliplatin and capecitabine as first-line treatment for advanced G/GEJ cancer. Patients with previously untreated, locally advanced or metastatic G/GEJ cancer were enrolled. All patients received HX008 3 mg/kg intravenously every 3 weeks, oxaliplatin 130 mg/m2 intravenously on day 1 every 3 weeks (up to 6 cycles), and capecitabine 1000 mg/m2 orally twice daily for 14 days continuous dosing followed by a 7-day break. The primary end point was the incidence of adverse events and serious adverse events. In total, 35 patients were enrolled. Median follow-up was 12.7 months. Most frequent (>10%) grade ≥3 treatment-related adverse events were anemia (27.5%), neutropenia (20%), thrombocytopenia (17.1%), leukopenia (17.1%) and fatigue (17.3%). Objective response rate was 60.0% (95% confidence interval [CI] 42.1–76.1%). Disease control rate was 77.1% (95% CI 59.9–89.6). Median time to response and duration of response were 1.4 months (range 1.3–2.9) and 12.3 months (range 1.4–17.9+), respectively. Median PFS was 9.2 months (95% CI 5.4-not reached). These results demonstrated that HX008 combined with oxaliplatin plus capecitabine was well tolerated and demonstrated encouraging efficacy as first-line treatment for advanced G/GEJ cancer. This study was registered in china, register number was CTR20181270.
- Published
- 2021
34. NB-UVB phototherapy in the treatment of anti-PD-1 inhibitor induced psoriasis: A case report
- Author
-
Vincent T. Ma, Johann E. Gudjonsson, Rajiv M. Patel, Charles S. Katzman, Ajjai Alva, and Phillip L. Palmbos
- Subjects
medicine.medical_specialty ,Side effect ,integumentary system ,business.industry ,NB-UVB phototherapy ,Immune checkpoint inhibitors ,Anti pd 1 ,General Engineering ,Cancer ,Narrow band uvb ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immune checkpoint inhibitor ,medicine.disease ,Dermatology ,Psoriasis ,medicine ,General Earth and Planetary Sciences ,business ,Adverse effect ,Anti-PD-1 inhibitor ,RC254-282 ,General Environmental Science - Abstract
Immune-mediated cutaneous reactions are a frequent occurrence in cancer patients treated with immune checkpoint inhibitors. A small subset of these adverse events are psoriasiform reactions. To date, there is no high-level evidence supporting the most appropriate management of this often debilitating dermatologic side effect. In many cases where topical therapy is ineffective, oral steroids are often prescribed which may lead to poorer outcomes in cancer patients. In this case report and review, we describe the clinical courses of two patients with anti-PD-1 inhibitor induced psoriasiform reactions who were treated with narrow band UVB phototherapy. We review the rationale, advantages, and disadvantages of this underutilized treatment as a therapeutic option to spare cancer patients from systemic immunosuppressive agents.
- Published
- 2020
35. Anti-PD-1, anti-VEGF, and temozolomide therapy in a patient with recurrent glioblastoma: a case report
- Author
-
Yanling Zhang, Wenwei Zuo, Pan Yang, and Can Chen
- Subjects
Adult ,Oncology ,Medicine (General) ,medicine.medical_specialty ,Bevacizumab ,Angiogenesis Inhibitors ,Case Report ,temozolomide ,bevacizumab ,Biochemistry ,03 medical and health sciences ,R5-920 ,0302 clinical medicine ,Internal medicine ,Overall survival ,Humans ,Medicine ,Anti vegf ,nivolumab ,Temozolomide ,Brain Neoplasms ,business.industry ,Recurrent glioblastoma ,Biochemistry (medical) ,Anti pd 1 ,Cell Biology ,General Medicine ,medicine.disease ,immunochemotherapy ,030220 oncology & carcinogenesis ,Female ,anti-angiogenesis ,Neoplasm Recurrence, Local ,Nivolumab ,business ,Glioblastoma ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Background Patients suffering from postoperative recurrent glioblastoma have an extremely unfavorable outcome because there are no proven therapeutic options. The median overall survival for those with relapsed glioblastoma after surgery is only 7.5 months. Case presentation: Between March 2015 and October 2019, a 44-year-old female patient with recurrent glioblastoma was treated by our medical team. After several failed rounds of therapy, the patient was subsequently treated with the anti-programmed death (PD)-1 antibody nivolumab, anti-vascular endothelial growth factor (VEGF) antibody bevacizumab, and cytotoxic agent temozolomide. Results The patient showed a sustainable complete response to the regimen. To date, there have been no serious toxic side effects. As of October 2019 (the last follow-up), the patient has been in complete remission for 17 months since recurrence. Conclusion The experience of this complicated case indicates the possible application of immune checkpoint inhibitors, anti-angiogenesis agents, and cytotoxic reagents for recurrent glioblastoma. The administration of this three-agent regimen appears safe and effective. However, further clinical trials are warranted.
- Published
- 2020
36. Mitochondria Determine Response to Anti-PD-1 Therapy: An Evidence-based Hypothesis
- Author
-
Farzad Taghizadeh-Hesary
- Subjects
chemistry.chemical_compound ,biology ,chemistry ,business.industry ,Programmed cell death 1 ,Anti pd 1 ,biology.protein ,other ,Medicine ,Pharmacology ,Mitochondrion ,business ,Adenosine triphosphate - Abstract
It has been demonstrated that a decrease in cellular adenosine triphosphate (c-ATP) causes cellular dysfunction. T-cells are not an exception. One of their roles is to properly detect and eliminate cancer cells. These processes occur at the expense of ATP. Therefore, it can be concluded that a decrease in c-ATP can defect T-cell function and promote cancer evolution. In this article, we provide a hypothesis to describe the correlation between the expression of PD-1 protein on T-cells and their c-ATP levels. Moreover, we present the possible predictive factors of Anti–PD(L)-1 therapy which has not yet been determined definitely.
- Published
- 2020
37. OR28-07 Increased BMI Is Associated With Anti PD-1/PD-L1-Induced Thyroid Immune-Related Adverse Events
- Author
-
Rena Pollack, Rivka Dresner-Pollak, Amit Ashash, and Avivit Cahn
- Subjects
Thyroid ,biology ,business.industry ,Endocrinology, Diabetes and Metabolism ,Anti pd 1 ,medicine.anatomical_structure ,Immune system ,No Longer a Pain in the Neck—Recent Insight into Thyroid Growth, Development, and Pathology ,PD-L1 ,Immunology ,biology.protein ,medicine ,Adverse effect ,business ,AcademicSubjects/MED00250 - Abstract
Background: Immune checkpoint inhibitors have revolutionized cancer therapy, however, are associated with immune related adverse events (irAEs). Obesity is a pro-inflammatory metabolic state that may play a role in the development of irAEs. Hypothesis: We hypothesized that likelihood of developing thyroid irAEs following anti-PD-1/L1 therapy increases with increasing body mass index (BMI). Methods: We retrospectively analyzed data of 187 cancer patients who initiated anti-PD-1/L1 at our institution between 01/2014-12/2018, had normal thyroid function tests at baseline and had baseline BMI data available. Results: Overall, 97 (52.2%) patients were with low-normal BMI (
- Published
- 2020
38. Efficacy and Safety of Anti–PD-1 Immunotherapy in Patients With Advanced NSCLC With BRAF, HER2, or MET Mutations or RET Translocation: GFPC 01-2018
- Author
-
Hélène Doubre, H. Janicot, Pierre Fournel, Catherine Dubos-Arvis, O. Bylicki, Pascal Assouline, Gilles Gonzales, Charles Ricordel, F. Vinas, Christos Chouaid, Florian Guisier, Stanislas Ropert, Maurice Pérol, Marie Bernardi, Lionel Falchero, Chantal Decroisette, R. Lamy, Jérôme Dauba, Service de pneumologie, oncologie thoracique et soins intensifs respiratoires [Rouen], Hôpital Charles Nicolle [Rouen]-CHU Rouen, Normandie Université (NU)-Normandie Université (NU)-Université de Rouen Normandie (UNIROUEN), Normandie Université (NU), Centre Régional de Lutte contre le Cancer François Baclesse [Caen] (UNICANCER/CRLC), UNICANCER-Tumorothèque de Caen Basse-Normandie (TCBN)-Normandie Université (NU), Institut Mondor de Recherche Biomédicale (IMRB), Institut National de la Santé et de la Recherche Médicale (INSERM)-IFR10-Université Paris-Est Créteil Val-de-Marne - Paris 12 (UPEC UP12), Service de pneumologie [Rennes] = Pneumology [Rennes], CHU Pontchaillou [Rennes], Service de Pneumologie [CHU Clermont-Ferrand], Pôle RHEUNNIRS [CHU Clermont-Ferrand], CHU Gabriel Montpied [Clermont-Ferrand], CHU Clermont-Ferrand-CHU Clermont-Ferrand-CHU Gabriel Montpied [Clermont-Ferrand], CHU Clermont-Ferrand-CHU Clermont-Ferrand, Institut de Cancérologie de la Loire Lucien Neuwirth, Centre Hospitalier Universitaire de Saint-Etienne (CHU de Saint-Etienne), Centre Léon Bérard [Lyon], and Normandie Université (NU)-UNICANCER-Tumorothèque de Caen Basse-Normandie (TCBN)
- Subjects
Male ,Proto-Oncogene Proteins B-raf ,0301 basic medicine ,Pulmonary and Respiratory Medicine ,Oncology ,medicine.medical_specialty ,Lung Neoplasms ,medicine.medical_treatment ,[SDV]Life Sciences [q-bio] ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Humans ,Medicine ,In patient ,Prospective Studies ,Prospective cohort study ,neoplasms ,Aged ,Retrospective Studies ,business.industry ,Proto-Oncogene Proteins c-ret ,Anti pd 1 ,Immunotherapy ,medicine.disease ,Confidence interval ,3. Good health ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,Cohort ,Adenocarcinoma ,Female ,business ,RET Translocation - Abstract
Introduction Immune-checkpoint inhibitor (ICI) efficacy in patients with NSCLC harboring molecular alterations remains poorly elucidated. This study was undertaken to determine ICI efficacy against BRAF-, HER2-, MET-, and RET-NSCLC in a real-world setting. Methods In this retrospective, multicenter study in ICI-treated BRAF-, HER2-, MET- or RET-NSCLCs, we analyzed clinical characteristics and outcomes: ICI-treatment duration, progression-free survival (PFS), objective response rate, duration of response, and overall survival (OS). Results There were 107 patients with NSCLC (mean age, 65.5 y) included from 21 centers: 37% were never-smokers, 54% were men, and 93% had adenocarcinoma. Among them, 44 had BRAF mutation (V600: 26), 23 had HER2 mutation, 30 had MET mutation, and nine had RET translocation. Programmed cell death ligand 1 (PD-L1) status was known for 70 patients and was greater than or equal to 1% in 34 patients. Before ICI, patients had received a median of one treatment line. Median duration of response, PFS, and OS were 15.4 (95% confidence interval [CI]: 12.6–not reached [NR]) months, 4.7 (95% CI: 2.3–7.4) months, and 16.2 (95% CI: 12.0–24.0) months, respectively, for the entire cohort. The response rates for BRAF-V600, BRAF–non-V600, HER2, MET, and RET-altered NSCLC were 26%, 35%, 27%, 36%, and 38%, respectively. For patients who were PD-L1 negative and those who were PD-L1 positive, PFS was 3.0 (95% CI: 1.2–NR) and 4.3 (95% CI: 2.1–8.5) months, respectively, and OS was 11.7 (95% CI: 4.1–NR) and 35.8 (95% CI: 9.0–35.2) months, respectively. Toxicities were reported in 28 patients (26%), including 11 patients (10%) with a grade greater than or equal to three. Conclusions In this real-world setting, ICI efficacy against patients with BRAF-, HER2-, MET-, or RET-NSCLC seemed close to that observed in unselected patients with NSCLC. Large prospective studies on these subsets of patients are needed.
- Published
- 2020
- Full Text
- View/download PDF
39. Positive progress: current and evolving role of immune checkpoint inhibitors in metastatic triple-negative breast cancer
- Author
-
Deanna McLeod, Christine Brezden-Masley, Anil Abraham Joy, Joy McCarthy, and Christine E. Simmons
- Subjects
business.industry ,medicine.medical_treatment ,Immune checkpoint inhibitors ,Anti pd 1 ,Breast cancer subtype ,Immunotherapy ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Breast cancer ,Oncology ,Atezolizumab ,medicine ,Cancer research ,business ,Triple-negative breast cancer - Abstract
Background: Triple-negative breast cancer (TNBC) represents an aggressive breast cancer subtype with historically poor overall outcomes, due primarily to a lack of effective targeted agents. Chemotherapy has been the primary treatment approach, although immune checkpoint inhibitors (ICIs) are currently being investigated to improve patient outcomes. This review examines the clinical implications of current evidence on the use of ICIs for the treatment of metastatic TNBC. Methods: Our systematic search identified two phase III and five phase I/II trials reporting on the efficacy of ICIs used as monotherapy or combined with chemotherapy for the treatment of metastatic TNBC. Results: The phase III IMpassion 130 trial showed a significant improvement in median progression-free survival in the intent-to-treat (net 1.7 months, p = 0.002) and PD-L1-positive populations (net 2.5 months, p Conclusions: Atezolizumab plus nab-paclitaxel represents a potential new first-line standard of care for patients with metastatic PD-L1-positive TNBC. Other ICIs used as monotherapy, or combined with chemotherapy for advanced TNBC, as well as their use for earlier stage disease, are areas of ongoing investigation.
- Published
- 2020
40. The anti–PD-1 antibody spartalizumab in combination with dabrafenib and trametinib in advanced BRAF V600–mutant melanoma : Efficacy and safety findings from parts 1 and 2 of the Phase III COMBI-i trial
- Author
-
Aisha Masood, Keith T. Flaherty, Reinhard Dummer, Erika Richtig, Hussein Abdul-Hassan Tawbi, Céleste Lebbé, Neha Pakhle, Caroline Robert, Paul Nathan, Naoya Yamazaki, Mario Mandalà, Dirk Schadendorf, Victoria Atkinson, Paolo A. Ascierto, Ana Arance, Antoni Ribas, Georgina V. Long, and Eduard Gasal
- Subjects
Trametinib ,Cancer Research ,business.industry ,Melanoma ,medicine.medical_treatment ,Anti pd 1 ,Mutant ,Medizin ,Dabrafenib ,medicine.disease ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,In patient ,business ,030215 immunology ,medicine.drug ,Advanced melanoma - Abstract
10028 Background: Treatment (tx) with checkpoint inhibitors or targeted therapy improves outcomes in patients (pts) with BRAF V600–mutant advanced melanoma; however, many pts subsequently progress and die. Preliminary evidence suggests that targeted therapy may enhance the impact of checkpoint inhibitors and improve efficacy compared with either treatment alone. Methods: COMBI-i is investigating first-line spartalizumab 400 mg every 4 wk + dabrafenib 150 mg twice daily + trametinib 2 mg once daily in pts with unresectable or metastatic BRAF V600–mutant melanoma (NCT02967692). We report efficacy and safety data from parts 1 (run-in cohort) and 2 (biomarker cohort), with a median follow-up of 24.3 mo. Response was assessed per RECIST v1.1. The randomized part 3 is ongoing. Results: 36 pts were enrolled (part 1: n = 9; part 2: n = 27); 20 (56%) had stage IV M1c disease and 15 (42%) had elevated lactate dehydrogenase (LDH) levels (≥ upper limit of normal). At the data cutoff (August 19, 2019), tx was ongoing in 10 pts (28%). The confirmed investigator-assessed objective response rate (ORR) was 78% (n = 28), with 16 complete responses (CRs; 44%) and 12 partial responses (33%). Median duration of response (DOR; 10/28 responders with events) was not reached (NR); 24-mo DOR rate was 53.4% (95% CI, 29%-73%). Median progression-free survival (PFS) was 22.7 mo; 24-mo PFS rate was 41.4% (95% CI, 23%-59%). At the cutoff, median overall survival (OS) was NR, with a 24-mo OS rate of 74.1% (95% CI, 56%-86%). In pts with elevated LDH, ORR was 67%, with 4 CRs (27%); median PFS was 10.7 mo (95% CI, 4.6-19.1 mo), and median OS was NR. The estimated 24-mo PFS and OS rates in these pts were 26.7% and 52.5%, respectively. All pts had ≥ 1 tx-related adverse event (TRAEs); 26 (72%) had grade ≥ 3 TRAEs. The most common grade ≥ 3 TRAEs were pyrexia (17%), increased lipase (11%), neutropenia (11%), increased blood creatine phosphokinase (8%), and increased γ-glutamyltransferase (8%). AEs leading to discontinuation of all 3 study drugs occurred in 6 pts (17%). All-causality grade ≥ 3 AEs requiring immunosuppressive medication occurred in 19 pts (53%). One pt died of cardiac arrest that was not considered tx related. Conclusions: The combination of spartalizumab + dabrafenib + trametinib resulted in high ORR and CR rates, with a high frequency of durable responses, including in patients with poor prognostic factors. Clinical trial information: NCT02967692.
- Published
- 2020
41. Overcoming Primary Resistance to PD-1 Inhibitor With Anti–PD-L1 Agent in Squamous-Cell NSCLC: Case Report
- Author
-
Daria Maria Filippini, Stefano Brocchi, Francesca Sperandi, Filippo Gustavo Dall'Olio, Alessandro Federico, Caterina Balacchi, Andrea Ardizzoni, Giuseppe Lamberti, Francesco Gelsomino, Gelsomino F., Di Federico A., Filippini D.M., Dall'Olio F.G., Lamberti G., Sperandi F., Balacchi C., Brocchi S., and Ardizzoni A.
- Subjects
Pulmonary and Respiratory Medicine ,Cancer Research ,biology ,business.industry ,medicine.medical_treatment ,Cell ,Anti pd 1 ,Immune checkpoint inhibitor rechallenge ,Immunotherapy ,medicine.disease ,medicine.anatomical_structure ,Nivolumab ,Oncology ,Atezolizumab ,Programmed cell death 1 ,Cancer research ,medicine ,biology.protein ,Lung cancer ,business - Abstract
Clinical Practice Points •Immune checkpoint inhibitors (ICI) have changed the treatment landscape of advanced non–small-cell lung cancer, as they have shown their superiority to chemotherapy in the first-line setting in tumors having programmed death ligand 1 (PD-L1) expression ≥ 50% and in patients with pretreated disease, regardless of PD-L1 expression. • The efficacy and safety of ICI rechallenge in those patients whose disease failed to respond to a prior treatment with these agents remain unclear. • We report the case of a 79-year-old woman, a smoker, with locally advanced, TP53-mutated squamous non–small-cell lung cancer, whose disease responded to an anti–PD-L1 agent despite having experienced disease progression during a prior anti–programmed cell death 1 treatment that followed first-line chemoradiotherapy. The anti–PD-L1 therapy was still ongoing after 9 months. • Our case suggests that ICI rechallenge could be safe and effective, even in a subpopulation of patients with disease that did not respond to prior ICI therapy.
- Published
- 2020
42. Impact of anti-PD-1 and anti-CTLA-4 on the HIV reservoir in vivo: AMC-095 Study
- Author
-
SharonR Lewin, Danielle N. Rigau, Thomas A Rasmussen, Rachel L. Rutishauser, Michael P. Busch, Shelly Lensing, Christine M. Durand, Sonia Bakkour, Dirk P. Dittmer, Surekha Tennakoon, Ashanti Dantanarayana, Steven G. Deeks, Ajantha Rhodes, Socheata Chea, and Laskhmi Rajdev
- Subjects
Epidemiology ,business.industry ,Immunology ,Anti pd 1 ,Public Health, Environmental and Occupational Health ,Human immunodeficiency virus (HIV) ,Anti ctla 4 ,medicine.disease_cause ,Microbiology ,QR1-502 ,Infectious Diseases ,In vivo ,Virology ,Medicine ,Public aspects of medicine ,RA1-1270 ,business - Published
- 2019
43. Title: Case of Hyperthyroidism in a Patient on Anti-PD-1 (Programmed Cell Death Receptor-1 Blocking Antibody) Therapy Caused by Destructive Thyroiditis Followed by Graves’ Disease
- Author
-
Mohsen Zena and Chandrika Reddy
- Subjects
Thyroid ,Programmed cell death ,endocrine system ,endocrine system diseases ,business.industry ,Endocrinology, Diabetes and Metabolism ,Graves' disease ,Anti pd 1 ,medicine.disease ,Thyroiditis ,Thyroid Disorders Case Report ,Blocking antibody ,Immunology ,medicine ,business ,Receptor ,hormones, hormone substitutes, and hormone antagonists ,AcademicSubjects/MED00250 - Abstract
Introduction: Immune checkpoint inhibitor (ICI) related thyroid dysfunction such as destructive thyroiditis and hypothyroidism can occur with Anti-PD-1 therapy. Graves’ disease (GD) however is considered rare. We report a case of isolated hyperthyroidism in a patient on Pembrolizumab, Anti-PD-1 presenting initially as destructive thyroiditis and later transitioned to GD. Case: 57 y.o lady was diagnosed with squamous cell carcinoma of cervix, stage 4 in 2016. She was noted to have asymptomatic goiter in 1997. PET scan in 02/2017 showed hypermetabolic activity in the right lobe nodule of 5.6 cm with maximum SUV of 9.8. Baseline TSH was in normal range in 04/2017. US guided fine needle biopsy of the nodule resulted as follicular neoplasm with 15-30%, risk of malignancy. Molecular test showed 40% risk of malignancy. Lobectomy was deferred. Pembrolizumab was initiated for advanced cervical cancer in 07/2018. Repeat PET in 10/2018 showed resolution of metastasis and right thyroid mass was stable. TSH remained in euthyroid range for 4 months until 11/ 2018. In 01/2019, her TSH was low and became undetectable in 03/2019 with high free T4 and total T3. She developed symptoms of hyperthyroidism. TSI was positive and TSHR Ab was negative. RAIU was low. No iodinated contrast study done prior to I123 scan. In 05/2019 to 08/2019, free T4 and total T3 was normal range with detectable but low TSH consistent with transient resolving thyroiditis. In 10/2019, thyroid hormones significantly increased with recurrence of hyperthyroidism. Both TSI and TSHR Ab were elevated confirming diagnosis of GD. Due to overt thyrotoxicosis, Methimazole (MMI) was initiated in 10/2019. She was briefly lost to follow up and stopped MMI. RAIU scan in 12 /2019 showed increased uptake consistent with GD. MMI was resumed in 04/2020. 06/2020 - 09/2020, MMI dose was titrated to achieve euthyroid state. In 09/25/2020, hypothyroidism was noted which improved with MMI dose reduction. Anti- TPO and Anti-Tg were negative. Pembrolizumab was stopped in 07/2020 due to dilated cardiomyopathy. She completed 2-year course of Pembrolizumab with advanced cervical cancer showing excellent response. Discussion: Observed thyroid dysfunction with ICI, is transient destructive thyroiditis followed by hypothyroidism, which can be permanent. Our patient had an initial low RAIU indicating destructive thyroiditis along with, mildly elevated TSI suggestive of superimposed autoimmune process. The elevated TSI predisposed to later occurrence of GD likely precipitated by Pembrolizumab. MMI provided therapeutic benefit. Conclusion: Less than 10 cases of GD with ICI and 5 cases of GD with Anti-PD- 1 have been reported. Our patient is a second case of persistent hyperthyroidism due to GD induced by Pembrolizumab. GD should be considered if hyperthyroidism persists or recurs in patients on ICI with destructive thyroiditis, which is mostly transient.
- Published
- 2021
44. Avelumab: clinical trial innovation and collaboration to advance anti-PD-L1 immunotherapy
- Author
-
D.S.A. Nuyten, Kevin M. Chin, and Vikram Chand
- Subjects
0301 basic medicine ,Oncology ,Drug ,medicine.medical_specialty ,medicine.medical_treatment ,media_common.quotation_subject ,MEDLINE ,Antibodies, Monoclonal, Humanized ,B7-H1 Antigen ,Avelumab ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,Internal medicine ,Neoplasms ,medicine ,Humans ,media_common ,Solid tumour ,Clinical Trials, Phase I as Topic ,Dose-Response Relationship, Drug ,business.industry ,Anti pd 1 ,Antibodies, Monoclonal ,Hematology ,Immunotherapy ,Industry Corner: Perspectives and Controversies ,Clinical trial ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunology ,business ,medicine.drug - Published
- 2017
45. Wong-type dermatomyositis during anti–PD-1 therapy
- Author
-
Wesley Y. Yu, Kanade Shinkai, Timothy H. McCalmont, and Jeffrey P. North
- Subjects
medicine.medical_treatment ,Immune checkpoint inhibitors ,erythema and urticaria ,Case Report ,Dermatology ,Colony stimulating factor 1 receptor ,immunology ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,Cancer immunotherapy ,Programmed cell death 1 ,medicine ,biology ,business.industry ,Anti pd 1 ,cancer and oncology ,Dermatomyositis ,medicine.disease ,inflammatory skin disease ,Pityriasis rubra ,drug reactions ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,therapy and treatment ,Nivolumab ,business ,CSF1R, colony-stimulating factor 1 receptor ,PD-1, programmed cell death 1 - Abstract
Checkpoint inhibitors used for cancer immunotherapy may also result in autoimmunity.1, 2 We present a case of Wong-type dermatomyositis complicating treatment with nivolumab, an inhibitor of the programmed cell death 1 (PD-1) receptor. Wong-type dermatomyositis is a rare variant of classic dermatomyositis with concurrent clinical and histopathologic findings mimicking pityriasis rubra pilaris.3 Our report of nivolumab-related Wong-type dermatomyositis expands the clinical spectrum of autoimmune diseases associated with checkpoint inhibitors.
- Published
- 2018
46. Results from a randomised phase I/II trial evaluating the safety and antitumour activity of anti-PD-1 (MEDI0680)/anti-PD-L1 (durvalumab) vs anti-PD-1 (nivolumab) alone in metastatic clear cell renal cell carcinoma (ccRCC)
- Author
-
L. Lewis, S.J. Welsh, H. Hu, Sjoukje F. Oosting, Ikbel Achour, Aaron R. Hansen, F.L. Walcott, A.A. Azad, Martin H. Voss, Jhanelle E. Gray, and Guided Treatment in Optimal Selected Cancer Patients (GUTS)
- Subjects
medicine.medical_specialty ,Durvalumab ,business.industry ,Disease progression ,Anti pd 1 ,Hematology ,EPIC ,Phase i ii ,Oncology ,Family medicine ,medicine ,Disease characteristics ,Nivolumab ,business ,Measurable Lesion - Abstract
Background MEDI0680 is a humanised IgG4κ anti-programmed cell death-1 (PD-1) mAb. We hypothesised that simultaneous blockade of PD-1:PD-L1/PD-L2 with MEDI0680 (M) and PD-1:PD-L1/CD80 with anti-PD-L1 mAb durvalumab (D) would improve efficacy vs blockade of the PD-1:PD-L1/PD-L2 pathway (with nivolumab; N) alone. M+D was well tolerated in the dose-escalation phase of a Phase I/II study in pts with advanced solid tumours, with an ORR of 33% (10/30; including 3/4 RCC pts). In the Phase II portion of the study, we compared M+D to N in a dose-expansion cohort of pretreated, immunotherapy (IO)-naive pts with metastatic ccRCC. Methods Eligible pts had received 1–3 prior therapy lines, no prior IO exposure and ≥1 measurable lesion. They were randomised 2:1 (stratified by MSKCC risk group and PD-L1 expression) to M 20 mg/kg IV + D 750 mg Q2W or N 240 mg IV Q2W until unacceptable toxicity or disease progression, for ≤2 years. Endpoints included investigator-assessed ORR by RECIST v1.1 (primary endpoint), PFS and safety (secondary). Sample size was ∼60 to detect a difference of 26.0% (ie, ORR = 47.5%, assuming ORR of 21.5% for N) with 76% power at a 1-sided significance level of 0.10. Results By Feb 24, 2019, 63 pts were randomised. Baseline pt/disease characteristics were generally well balanced, but more pts on N had favourable MSKCC risk (7/21; 33.3%) vs M+D (10/42; 23.8%). ORR was 14.3% (6/42; 2 CR, 4 PR; plus 2 unconfirmed PR) vs 19.0% (4/21; 4 PR, 0 unconfirmed) for M+D and N, respectively. There was no difference between arms in ORR by PD-L1 expression ( Conclusions Efficacy was similar with combined M+D and N monotherapy in pts with TKI-pretreated, IO-naive, metastatic ccRCC, but more pts discontinued M+D due to TRAEs. Clinical trial identification NCT02118337. Editorial acknowledgement Aaron Korpal, PhD, of Cirrus Communications (Macclesfield, UK), an Ashfield company, funded by AstraZeneca. Legal entity responsible for the study AstraZeneca. Funding AstraZeneca. Disclosure M.H. Voss: Research grant / Funding (institution): BMS; Research grant / Funding (institution): Genentech; Honoraria (self): Eisai; Honoraria (self): Exelixis; Honoraria (self): Pfizer; Honoraria (self): Novartis; Honoraria (self): Calithera; Honoraria (self): Corvus. A.A. Azad: Honoraria (self), Advisory / Consultancy: Janssen; Honoraria (self), Advisory / Consultancy, Research grant / Funding (institution), Non-remunerated activity/ies: Astellas; Honoraria (self), Advisory / Consultancy: Novartis; Research grant / Funding (institution), Non-remunerated activity/ies: Merck Serono; Honoraria (self), Advisory / Consultancy: Tolmar; Honoraria (self), Advisory / Consultancy, Non-remunerated activity/ies: Amgen; Honoraria (self), Advisory / Consultancy: Pfizer; Honoraria (self): Bayer; Honoraria (self), Advisory / Consultancy: Telix Pharmaceuticals; Honoraria (self), Advisory / Consultancy: Bristol-Myers Squibb; Honoraria (self), Advisory / Consultancy: Sanofi. A.R. Hansen: Advisory / Consultancy, Research grant / Funding (institution): Genentech/Roche; Advisory / Consultancy, Research grant / Funding (institution): Merck; Advisory / Consultancy, Research grant / Funding (institution): GSK; Advisory / Consultancy, Research grant / Funding (institution): Bristol-Myers Squibb; Advisory / Consultancy, Research grant / Funding (institution): Novartis; Research grant / Funding (institution): Boston Biomedical; Advisory / Consultancy, Research grant / Funding (institution): Boehringer Ingelheim; Advisory / Consultancy, Research grant / Funding (institution): AstraZeneca. J.E. Gray: Honoraria (self), Advisory / Consultancy, Research grant / Funding (institution): AstraZeneca; Honoraria (self), Advisory / Consultancy, Research grant / Funding (institution): Genentech; Honoraria (self), Advisory / Consultancy: Celgene; Honoraria (self), Advisory / Consultancy: Takeda; Honoraria (self), Advisory / Consultancy: Janssen; Honoraria (self), Advisory / Consultancy: Eli Lilly; Honoraria (self), Advisory / Consultancy: Triptych Health Partners; Research grant / Funding (institution): Array; Research grant / Funding (institution): Merck; Research grant / Funding (institution): Epic Sciences; Research grant / Funding (institution): BMS; Research grant / Funding (institution): BI; Research grant / Funding (institution): Trovagene; Research grant / Funding (institution): Loxo; Research grant / Funding (institution): Blueprint; Research grant / Funding (institution): Novartis. I. Achour: Shareholder / Stockholder / Stock options, Full / Part-time employment: AstraZeneca. H. Hu: Full / Part-time employment: AstraZeneca. L. Lewis: Travel / Accommodation / Expenses, Full / Part-time employment: AstraZeneca. F.L. Walcott: Shareholder / Stockholder / Stock options, Full / Part-time employment: AstraZeneca. S.F. Oosting: Research grant / Funding (institution): Celldex; Research grant / Funding (institution): Novartis. All other authors have declared no conflicts of interest.
- Published
- 2019
47. Retroperitoneal fibrosis in on-going anti-PD-1 immunotherapy detected with [18F]-FDG PET/CT
- Author
-
Anne Laure Giraudet, Thomas Mognetti, A. Moreau, David Kryza, Laboratoire Traitement et Communication de l'Information (LTCI), Télécom ParisTech-Institut Mines-Télécom [Paris] (IMT)-Centre National de la Recherche Scientifique (CNRS), Laboratoire d'automatique, de génie des procédés et de génie pharmaceutique (LAGEPP), Université Claude Bernard Lyon 1 (UCBL), and Université de Lyon-Université de Lyon-École Supérieure Chimie Physique Électronique de Lyon-Centre National de la Recherche Scientifique (CNRS)
- Subjects
medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Anti pd 1 ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,General Medicine ,Immunotherapy ,Retroperitoneal fibrosis ,[SDV.SP.PG]Life Sciences [q-bio]/Pharmaceutical sciences/Galenic pharmacology ,[SDV.SP.MED]Life Sciences [q-bio]/Pharmaceutical sciences/Medication ,medicine ,Radiology, Nuclear Medicine and imaging ,Fdg pet ct ,Radiology ,medicine.symptom ,business ,ComputingMilieux_MISCELLANEOUS ,Positron Emission Tomography-Computed Tomography - Abstract
International audience
- Published
- 2019
- Full Text
- View/download PDF
48. Efficacy of anti-PD-1 antibody SHR-1210 as second-line treatment in hepatocellular carcinoma patient with sorafenib resistance
- Author
-
Yaqin Zhao, Hong Zhu, Xi Yang, and Cheng Yi
- Subjects
Oncology ,Sorafenib ,Male ,medicine.medical_specialty ,Poor prognosis ,Carcinoma, Hepatocellular ,Lung Neoplasms ,Programmed Cell Death 1 Receptor ,SHR-1210 ,Placebo ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Internal medicine ,Regorafenib ,medicine ,Humans ,030212 general & internal medicine ,Clinical Case Report ,HCC ,late onset ,neoplasms ,Second line treatment ,business.industry ,Anti pd 1 ,Liver Neoplasms ,General Medicine ,Middle Aged ,medicine.disease ,digestive system diseases ,Sorafenib resistance ,Treatment Outcome ,chemistry ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,second line ,alpha-Fetoproteins ,business ,excellent effect ,medicine.drug ,Research Article - Abstract
Rationale: Hepatocellular carcinoma (HCC), one of the most common cancers worldwide, is an aggressive tumor with very poor prognosis. Regorafenib was the first agent to show a survival benefit over placebo in patients who showed progression while being treated with sorafenib, but it remains an unsatisfactory agent owing to its serious side effects. Therefore, more efficient and milder therapies are needed. Patient concerns: Herein, we report a patient with advanced HCC with many lung metastases who showed progression during sorafenib treatment. Diagnoses: HCC with lung metastases (stage IVB). Interventions: SHR-1210 alone was used as second-line treatment. Outcomes: Although the lung metastases did not decrease 3 months after the treatment, they decreased significantly at 6 months after the treatment and partially disappeared. The tumor response indicated partial response. Furthermore, all of the lung metastases continued to decrease at about 17 months after treatment. The alpha-fetoprotein levels showed a similar trend. After a follow up of 19 months, the patient remains in good health. Lessons: SHR-1210 alone as a second-line treatment for a patient with HCC showed excellent antitumor effects. We think that SHR-1210 may exert its antitumor effects through a late-onset model, which persist for a long time. The side effects were mild and well tolerated.
- Published
- 2019
49. SAT-584 Pembrolizumab-Related Graves' Disease: A Rare Adverse Effect of an Anti-PD-1 Antibody Cancer Immunotherapy
- Author
-
Garima Narayen and David C. Lieb
- Subjects
Oncology ,Thyroid ,medicine.medical_specialty ,business.industry ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Graves' disease ,Anti pd 1 ,Pembrolizumab ,Thyroid Case Reports: Hypothyroidism, Hyperthyroidism, Goiter and Laboratory Assessment ,medicine.disease ,Cancer immunotherapy ,Internal medicine ,medicine ,Adverse effect ,business - Abstract
Background: Immune checkpoint inhibitors (anti-PD-1 and anti-CTLA-4 antibodies) have emerged as important anticancer therapies and are sometimes associated with thyrotoxicosis. With anti-PD-1 antibody therapy, most often this is related to destructive thyroiditis. We present a rare case of Graves’ disease related to pembrolizumab. Clinical Case: A 37‐year old woman presented with a 1-week history of lightheadedness, fatigue and nausea. She had a history of grade 3 diffuse oligodendroglioma diagnosed during evaluation for focal seizures three years ago. She had completed chemotherapy with temozolomide and then was found to have extension of the tumor into the right anterior temporal lobe. She underwent tumor resection and began pembrolizumab treatment shortly thereafter. She completed two infusions of pembrolizumab at the time of her presentation with the above symptoms. In the ED she was noted to be orthostatic and had a HR of 110 bpm. Labs were consistent with thyrotoxicosis: elevated FT3 of 10.5 pg/mL (2.3-4.2), FT4 3.7 ng/dL (0.9-1.8) and a suppressed TSH of < 0.01 mcU/mL. She was treated with IVF and started on propranolol 10 mg three times daily. A thyroid ultrasound revealed a hyperemic, diffusely heterogenous gland. Antibody testing demonstrated an elevated TSI of 10.8 IU/L (< 0.55), confirming a diagnosis of Graves’ disease. She was subsequently started on methimazole 10 mg twice daily and had improvement of both her symptoms and biochemical indices over several weeks. Up to 5% of patients receiving anti-PD-1 antibody immunotherapy develop thyrotoxicosis, with most reported patients having destructive thyroiditis.1 To our knowledge this is one of the first reports of thyrotoxicosis due to Graves’ disease related to anti-PD-1 antibody therapy. Interestingly there are two reports in the literature of Graves’ ophthalmopathy without thyrotoxicosis (one with pembrolizumab therapy alone and another with the combination of durvalumab and tremelimumab therapy).2,3 Our case demonstrates the importance of a thorough evaluation for the cause of thyrotoxicosis in patients receiving anti-PD-1 antibody therapy, as a prompt diagnosis allows for etiology-targeted management. Conclusion: This is the one of the first case reports demonstrating Graves’ disease as a specific thyroid-related endocrinopathy linked to PD-1 antagonist use. Clinicians must consider Graves’ disease as a potential cause of thyrotoxicosis in patients taking these medications. References: (1) Ferrari SM, Fallahi P, et al. Rev in Endocr and Metab Dis. 2018; Sep 21. Epub ahead of print. (2) Park ESY, Rabinowits G, et al. J of AAPOS. 2018;22(4):310-312 (3) Sabini E, Sframeli A, Marino M. J of Endocr Investig. 2018;41:877-878.
- Published
- 2019
50. Long-term survival with anti-PD-1-based immunotherapy, but what is the best approach?
- Author
-
Mark R. Middleton
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Melanoma ,Anti pd 1 ,MEDLINE ,Ipilimumab ,Immunotherapy ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,030220 oncology & carcinogenesis ,Internal medicine ,Long term survival ,medicine ,030212 general & internal medicine ,Nivolumab ,business ,medicine.drug - Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.