Phase IA/IB Study of Single-Agent Tislelizumab, an Investigational Anti

Total Page:16

File Type:pdf, Size:1020Kb

Phase IA/IB Study of Single-Agent Tislelizumab, an Investigational Anti Open access Original research J Immunother Cancer: first published as 10.1136/jitc-2019-000453 on 14 June 2020. Downloaded from Phase IA/IB study of single-agent tislelizumab, an investigational anti- PD-1 antibody, in solid tumors Jayesh Desai,1 Sanjeev Deva,2 Jong Seok Lee,3 Chia- Chi Lin,4 Chia- Jui Yen,5 Yee Chao,6 Bhumsuk Keam,7 Michael Jameson,8 Ming- Mo Hou,9 Yoon- Koo Kang,10 Ben Markman,11 Chang- Hsien Lu,12 Kun- Ming Rau,13 Kyung- Hun Lee,7 Lisa Horvath,14,15 Michael Friedlander,16 Andrew Hill,17 Shahneen Sandhu,1 Paula Barlow,2 Chi- Yuan Wu,18 Yun Zhang,19 Liang Liang,19 John Wu,18 Virginia Paton,18 Michael Millward20 To cite: Desai J, Deva S, ABSTRACT were observed in heavily pretreated patients with Lee JS, et al. Phase IA/IB study Background The programmed cell death-1/programmed advanced solid tumors, supporting the evaluation of of single- agent tislelizumab, cell death ligand-1 (PD-1/PD-L1) axis plays a central role tislelizumab 200 mg every 3 weeks, as monotherapy and in an investigational anti- PD-1 in suppressing antitumor immunity; axis dysregulation combination therapy, for the treatment of solid tumors and antibody, in solid tumors. can be used by cancer cells to evade the immune system. hematological malignancies. Journal for ImmunoTherapy Trial registration number NCT02407990. of Cancer 2020;8:e000453. Tislelizumab, an investigational monoclonal antibody doi:10.1136/jitc-2019-000453 with high affinity and binding specificity for PD-1, was engineered to minimize binding to FcγR on macrophages to limit antibody- dependent phagocytosis, a potential ► Additional material is INTRODUCTION published online only. To view, mechanism of resistance to anti-PD-1 therapy. The aim The programmed cell death-1/programmed please visit the journal online of this phase IA/IB study was to investigate the safety/ cell death ligand-1 (PD-1/PD- L1) axis plays (http:// dx. doi. org/ 10. 1136/ jitc- tolerability, antitumor effects and optimal dose and a central role in suppressing antitumor 2019- 000453). schedule of tislelizumab in patients with advanced solid immunity; dysregulation of the PD-1/PD-L1 tumors. axis can be used by cancer cells to evade the Accepted 23 April 2020 Methods Patients (aged ≥18 years) enrolled in phase 1 2 IA received intravenous tislelizumab 0.5, 2, 5 or 10 mg/ immune system. PD- L1 is an immune check- kg every 2 weeks; 2 or 5 mg/kg administered every point protein that is often overexpressed on 2 weeks or every 3 weeks; or 200 mg every 3 weeks; the surface of tumor and immune cells in http://jitc.bmj.com/ 3 4 patients in phase IB received 5 mg/kg every 3 weeks. the tumor microenvironment. PD-1, the Primary objectives were to assess tislelizumab’s safety/ cell receptor for PD-L1, is mainly expressed tolerability profile by adverse event (AE) monitoring and in activated T cells.5 An increase in PD-1 antitumor activity using RECIST V.1.1. PD- L1 expression expression in the tumor microenvironment was assessed retrospectively with the VENTANA PD-L1 has been reported in many cancer types.6–8 (SP263) Assay. Increased expression of PD-1 and PD-L1 is on September 30, 2021 by guest. Protected copyright. Results Between May 2015 and October 2017, 451 often associated with poor survival but may patients (n=116, IA; n=335, IB) were enrolled. Fatigue be predictive of anti-PD-1/PD- L1 antitumor (28%), nausea (25%) and decreased appetite (20%) were 9–11 the most commonly reported AEs. Most AEs were grade activity. 1–2 severity; anemia (4.9%) was the most common grade Tislelizumab is an investigational human- 3–4 AE. Treatment-rela ted AEs led to discontinuation ized IgG4 monoclonal antibody with high 12 in 5.3% of patients. Grade 5 AEs were reported in 14 affinity and binding specificity for PD-1. patients; 2 were considered related to tislelizumab. Tislelizumab was engineered to minimize Pneumonitis (2%) and colitis (1%) were the most common binding to FcγR on macrophages in order © Author(s) (or their serious tislelizumab-rela ted AEs. As of May 2019, 18% to limit antibody-dependent phagocytosis, employer(s)) 2020. Re- use of patients achieved a confirmed objective response in a potential mechanism of resistance to anti- permitted under CC BY-NC. No phase IA and 12% in phase IB; median follow-up duration PD-1 therapy.1 Preclinical data suggest tisleli- commercial re- use. See rights was 13.6 and 7.6 months, respectively. Pharmacokinetics, and permissions. Published by zumab does not bind to FcγRI, whereas BMJ. safety and antitumor activity obtained from both phase IA and IB determined the tislelizumab recommended dose; other anti- PD-1 antibodies bind to FcγRI in a For numbered affiliations see ultimately, tislelizumab 200 mg intravenous every 3 weeks manner consistent with human IgG4 antibody end of article. 12 was the dose and schedule recommended to be taken into affinity. Furthermore, in cell- based assays, Correspondence to subsequent clinical trials. tislelizumab enhanced the functional activity Dr Jayesh Desai; Conclusions Tislelizumab monotherapy demonstrated an of human T cells and pre- activated primary Jayesh. Desai@ petermac. org acceptable safety/tolerability profile. Durable responses peripheral blood mononuclear cells.12 Desai J, et al. J Immunother Cancer 2020;8:e000453. doi:10.1136/jitc-2019-000453 1 Open access J Immunother Cancer: first published as 10.1136/jitc-2019-000453 on 14 June 2020. Downloaded from KEY PHASE IA, PART 1 PHASE IA, PART 2 PHASE IA, PART 3 OBJECTIVES Dose Escalations Schedule Expansion* Fixed Dose Expansion†‡ PHASE IA 0.5 mg/kg Q2W, n=3 2 mg/kg Q2W, n=20 Safety, 2 mg/kg Q2W, n=6 2 mg/kg Q3W, n=21 200 mg Q3W RP2D, and n=13 preliminary 5 mg/kg Q2W, n=6 5 mg/kg Q2W, n=20 efficacy 10 mg/kg Q2W, n=7 5 mg/kg Q3W, n=20 5 mg/kg Q3W PHASE IB: Indication Expansion Expansion in ~330 patients with multiple tumor types ARM 1: n=50 ARM 2: n=20 ARM 3: n=50 ARM 4: n=50 Non-small cell Hepatocellular PHASE IB Ovarian cancer Gastric cancer lung cancer carcinoma Efficacy and safety in ARM 5: n=20 ARM 6: n=50 ARM 7: n=20 ARM 8: n=20 Head and neck Triple-negative Esophageal carcinoma Cholangiocarcinoma multiple squamous cell carcinoma breast cancer tumor types ARM 9: n=50 Renal cell carcinoma, urothelial cancer, melanoma, Merkel cell carcinoma, sarcoma, gastrointestinal stromal tumor, or cutaneous squamous cell carcinoma. Or any other metastatic microsatellite instability-high or mismatch repair deficient solid tumors, such as CRC or pancreatic cancer Figure 1 Study design. CRC, colorectal cancer; Q3W, every 3 weeks. This first- in- human (FIH), dose- escalation/dose- diarrhea, nausea and vomiting; asymptomatic biochem- expansion study assessed the safety/tolerability, pharma- ical abnormalities that improve to grade ≤2 within 3 days cology and clinical activity of tislelizumab in patients with of institution of supportive care and grade 2 ophthalmo- advanced solid tumors. The primary objective was to eval- logical toxicities. Hematological toxicities meeting DLT uate the safety and tolerability of tislelizumab (phase IA), criteria include febrile neutropenia (defined as absolute as well as the antitumor response (phase IB). Secondary neutrophil count <1000/mm3 with a single temperature end points included determining the maximum toler- of 38.3°C or a sustained temperature of 38°C for >1 hour); http://jitc.bmj.com/ ated dose (MTD) and the optimal dose and treatment grade 4 thrombocytopenia, grade 4 anemia or grade 4 regimen. Confirmed objective response rate (ORR) to neutropenia lasting >7 days; grade 3 thrombocytopenia tislelizumab by PD- L1 status was an exploratory end point. with bleeding and grade 3 neutropenic infection. No additional patients were required if a DLT was observed METHODS in one of six patients. Dose escalation continued until on September 30, 2021 by guest. Protected copyright. Study design and treatment administration two out of six patients in each dose cohort experienced a The design of this phase IA/IB study is detailed in DLT. If an MTD could not be established after evaluation figure 1. Phase IA comprised three parts. Part 1 was a of all planned doses, then the dose for subsequent studies dose- escalation/dose- finding phase that followed a modi- would be determined based on safety and pharmacoki- fied 3+3 design. Four weight-based dose levels (0.5, 2, netic (PK) profile from aggregated data across the study. 5 and 10 mg/kg every 2 weeks) were assessed for dose- Part 2 of phase IA was a schedule expansion that evaluated limiting toxicity (DLT). If a DLT was observed in the first tislelizumab at 2 and 5 mg/kg every 2 weeks and every 3 three patients completing one cycle of treatment at a weeks. Part 3 of phase IA (fixed-dose expansion) evalu- given dose level, three additional patients were enrolled. ated tislelizumab 200 mg administered every 3 weeks. The A DLT was defined as an adverse event (AE) occurring phase IB component of this study was an expansion phase in the first 28- day cycle that met the predefined criteria of tislelizumab at 5 mg/kg every 3 weeks in nine disease- based on grading per NCI-CTCAE V.4.03. DLT criteria included any grade ≥4 non-hematological toxicity or specific cohorts. The initial administration of tislelizumab grade 4 laboratory anomaly irrespective of duration; was administered via a 60 min infusion, if tolerated, the any grade ≥3 immune- related AE (irAE), grade 3 tumor infusion was shortened to 30 min for all subsequent flare (ie, local pain, irritation or rash localized at sites of administrations.
Recommended publications
  • (PDCO) Minutes of the Meeting on 29 January - 01 February 2019
    1 March 2019 EMA/PDCO/56017/2019 Inspections, Human Medicines Pharmacovigilance and Committees Division Paediatric Committee (PDCO) Minutes of the meeting on 29 January - 01 February 2019 Chair: Dirk Mentzer – Vice-Chair: Koenraad Norga 29 January 2019, 14:00- 19:00, room 3A 30 January 2019, 08:30- 19:00, room 3A 31 January 2019, 08:30- 19:00, room 3A 01 February 2019, 08:30- 13:00, room 3A Disclaimers Some of the information contained in this set of minutes is considered commercially confidential or sensitive and therefore not disclosed. With regard to intended therapeutic indications or procedure scopes listed against products, it must be noted that these may not reflect the full wording proposed by applicants and may also vary during the course of the review. Additional details on some of these procedures will be published in the PDCO Committee meeting reports (after the PDCO Opinion is adopted), and on the Opinions and decisions on paediatric investigation plans webpage (after the EMA Decision is issued). Of note, this set of minutes is a working document primarily designed for PDCO members and the work the Committee undertakes. Further information with relevant explanatory notes can be found at the end of this document. Note on access to documents Some documents mentioned in the minutes cannot be released at present following a request for access to documents within the framework of Regulation (EC) No 1049/2001 as they are subject to on-going procedures for which a final decision has not yet been adopted. They will become public when adopted or considered public according to the principles stated in the Agency policy on access to documents (EMA/127362/2006).
    [Show full text]
  • Mirati's Clinical Programs
    NASDAQ: MRTX Targeting the genetic and immunological drivers of cancer Corporate Presentation August 2019 1 Safe Harbor Statement Certain statements contained in this presentation, other than statements of fact that are independently verifiable at the date hereof, are "forward-looking" statements, within the meaning of the Private Securities Litigation Reform Act of 1955, that involve significant risks and uncertainties. Forward looking statements can be identified by the use of forward looking words such as “believes,” “expects,” “hopes,” “may,” “will,” “plan,” “intends,” “estimates,” “could,” “should,” “would,” “continue,” “seeks,” “pro forma,” or “anticipates,” or other similar words (including their use in the negative), or by discussions of future matters such as the development of current or future product candidates, timing of potential development activities and milestones, business plans and strategies, possible changes in legislation and other statements that are not historical. Forward-looking statements are based on current expectations of management and on what management believes to be reasonable assumptions based on information currently available to them, and are subject to risks and uncertainties. Such risks and uncertainties may cause actual results to differ materially from those anticipated in the forward-looking statements. Such risks and uncertainties include without limitation potential delays in development timelines, negative clinical trial results, reliance on third parties for development efforts, changes in the competitive landscape, changes in the standard of care, as well as other risks detailed in Mirati's recent filings on Forms 10-K and 10-Q with the U.S. Securities and Exchange Commission. Except as required by law, Mirati undertakes no obligation to update any forward-looking statements to reflect new information, events or circumstances, or to reflect the occurrence of unanticipated events.
    [Show full text]
  • Immune Checkpoint Inhibitor Combinations Current Efforts And
    Drug Resistance Updates 45 (2019) 13–29 Contents lists available at ScienceDirect Drug Resistance Updates journal homepage: www.elsevier.com/locate/drup Immune checkpoint inhibitor combinations: Current efforts and important aspects for success T ⁎ Edo Kon, Itai Benhar Department of Molecular Microbiology and Biotechnology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 69978, Israel ARTICLE INFO ABSTRACT Keywords: Immune checkpoint inhibitors (ICI) have emerged as a remarkable treatment option for diverse cancer types. Resistance Currently, ICIs are approved for an expanding array of cancer indications. However, the majority of patients still Immunotherapy do not demonstrate a durable long-term response following ICI therapy. In addition, many patients receiving ICI Clinic therapy develop immune-related adverse events (irAEs) affecting a wide variety of organs. To increase the CAR-T percentage of patients who benefit from ICI therapy and to reduce the occurrence of irAEs, there is an ongoing Radiation therapy effort to combine current ICIs with novel checkpoints inhibitors or other therapeutic approaches to achieve a Chemotherapy ff RNA cancer vaccines synergistic e ect which is larger than the sum of its parts. Angiogenesis In this review we highlight the essential factors for more effective ICI combinations. We describe how the Tumor microenvironment design of these strategies should be driven by the tumor's immunological context. We analyze current combi- nation strategies and describe how they can be improved to unleash the immune system's full anti-cancer po- tential as well as convert immunologically "cold" tumors into "hot" ones.
    [Show full text]
  • Immune-Checkpoint Blockade Therapy in Lymphoma
    International Journal of Molecular Sciences Review Immune-Checkpoint Blockade Therapy in Lymphoma Ayumi Kuzume 1,2, SungGi Chi 1 , Nobuhiko Yamauchi 1 and Yosuke Minami 1,* 1 Department of Hematology, National Cancer Center Hospital East, Kashiwa 277–8577, Japan; [email protected] (A.K.); [email protected] (S.C.); [email protected] (N.Y.) 2 Department of Hematology, Kameda Medical Center, Kamogawa 296–8602, Japan * Correspondence: [email protected]; Tel.: +81-4-7133-1111; Fax: +81-7133-6502 Received: 11 June 2020; Accepted: 28 July 2020; Published: 30 July 2020 Abstract: Tumor cells use immune-checkpoint pathways to evade the host immune system and suppress immune cell function. These cells express programmed cell-death protein 1 ligand 1 (PD-L1)/PD-L2, which bind to the programmed cell-death protein 1 (PD-1) present on cytotoxic T cells, trigger inhibitory signaling, and reduce cytotoxicity and T-cell exhaustion. Immune-checkpoint blockade can inhibit this signal and may serve as an effective therapeutic strategy in patients with solid tumors. Several trials have been conducted on immune-checkpoint inhibitor therapy in patients with malignant lymphoma and their efficacy has been reported. For example, in Hodgkin lymphoma, immune-checkpoint blockade has resulted in response rates of 65% to 75%. However, in non-Hodgkin lymphoma, the response rate to immune-checkpoint blockade was lower. In this review, we evaluate the biology of immune-checkpoint inhibition and the current data on its efficacy in malignant lymphoma, and identify the cases in which the treatment was more effective.
    [Show full text]
  • Clinical Potential of Kinase Inhibitors in Combination with Immune Checkpoint Inhibitors for the Treatment of Solid Tumors
    International Journal of Molecular Sciences Review Clinical Potential of Kinase Inhibitors in Combination with Immune Checkpoint Inhibitors for the Treatment of Solid Tumors Ryuhjin Ahn 1 and Josie Ursini-Siegel 2,3,4,5,* 1 Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; [email protected] 2 Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada 3 Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC H3T 1E2, Canada 4 Department of Experimental Medicine, McGill University, Montréal, QC H3A 0G4, Canada 5 Department of Oncology, McGill University, 546 Pine Avenue West, Montréal, QC H2W 1S6, Canada * Correspondence: [email protected]; Tel.: +514-340-8222 (ext. 26557); Fax: +514-340-7502 Abstract: Oncogenic kinases contribute to immunosuppression and modulate the tumor microenvi- ronment in solid tumors. Increasing evidence supports the fundamental role of oncogenic kinase signaling networks in coordinating immunosuppressive tumor microenvironments. This has led to numerous studies examining the efficacy of kinase inhibitors in inducing anti-tumor immune responses by increasing tumor immunogenicity. Kinase inhibitors are the second most common FDA-approved group of drugs that are deployed for cancer treatment. With few exceptions, they inevitably lead to intrinsic and/or acquired resistance, particularly in patients with metastatic disease when used as a monotherapy. On the other hand, cancer immunotherapies, including immune checkpoint inhibitors, have revolutionized cancer treatment for malignancies such as melanoma and Citation: Ahn, R.; Ursini-Siegel, J. lung cancer. However, key hurdles remain to successfully incorporate such therapies in the treatment Clinical Potential of Kinase Inhibitors of other solid cancers.
    [Show full text]
  • Natural Killer Cell-Based Immunotherapy for Acute Myeloid Leukemia
    Xu and Niu J Hematol Oncol (2020) 13:167 https://doi.org/10.1186/s13045-020-00996-x REVIEW Open Access Natural killer cell-based immunotherapy for acute myeloid leukemia Jing Xu and Ting Niu* Abstract Despite considerable progress has been achieved in the treatment of acute myeloid leukemia over the past decades, relapse remains a major problem. Novel therapeutic options aimed at attaining minimal residual disease-negative complete remission are expected to reduce the incidence of relapse and prolong survival. Natural killer cell-based immunotherapy is put forward as an option to tackle the unmet clinical needs. There have been an increasing num- ber of therapeutic dimensions ranging from adoptive NK cell transfer, chimeric antigen receptor-modifed NK cells, antibodies, cytokines to immunomodulatory drugs. In this review, we will summarize diferent forms of NK cell-based immunotherapy for AML based on preclinical investigations and clinical trials. Keywords: Acute myeloid leukemia, Natural killer cells, Immunotherapy, Adoptive NK cell transfer, Chimeric antigen receptor-modifed NK cells, Antibodies, Cytokines Background cells and substances in the immune system play pivotal Acute myeloid leukemia (AML) is a clinically and geneti- roles in detecting and destroying pathogen-infected or cally heterogeneous disease with unsatisfactory out- neoplastically transformed cells. But they become less comes. Over the last few years, considerable progress has potent in cancer elimination when malignant cells dis- been achieved in the treatment of AML with the devel- play the loss of antigenicity and/or immunogenicity and opment and implementation of new drugs [1, 2]. How- are surrounded by an immunosuppressive microenvi- ever, allogeneic hematopoietic cell transplantation (HCT) ronment [6].
    [Show full text]
  • Immune Checkpoint Inhibitors for the Treatment of Bladder Cancer
    cancers Review Immune Checkpoint Inhibitors for the Treatment of Bladder Cancer Antonio Lopez-Beltran 1,*,† , Alessia Cimadamore 2,† , Ana Blanca 3, Francesco Massari 4 , Nuno Vau 5, Marina Scarpelli 2, Liang Cheng 6 and Rodolfo Montironi 2,* 1 Unit of Anatomic Pathology, Department of Morphological Sciences, Cordoba University Medical School, 14004 Cordoba, Spain 2 Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60126 Ancona, Italy; [email protected] (A.C.); [email protected] (M.S.) 3 Maimonides Biomedical Research Institute of Cordoba, Department of Urology, University Hospital of Reina Sofia, 14004 Cordoba, Spain; [email protected] 4 Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; [email protected] 5 Medical Oncology, Champalimaud Clinical Center, 1400-038 Lisbon, Portugal; [email protected] 6 Department of Pathology and Laboratory Medicine, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; [email protected] * Correspondence: [email protected] or [email protected] (A.L.-B.); [email protected] (R.M.); Tel.: +34-9-5721-8992 (A.L.-B.); +39-0-71-596-4830 (R.M.); Fax: +34-9-5721-8229 (A.L.-B.) † These authors contributed equally to the work. Simple Summary: In this review, we examined relevant clinical trial results with immune check- point inhibitors in patients with metastatic urothelial cancer. We also focused on the potential of immunotherapy in the adjuvant and neoadjuvant setting or as part of drug combinations. Finally, we briefly review the current landscape of biomarkers of response to immune checkpoint inhibitors, such as programmed death-ligand 1 (PD-L1) expression, tumor mutation burden, molecular subtypes Citation: Lopez-Beltran, A.; of bladder cancer, and immune-gene expression profiling.
    [Show full text]
  • Immune Checkpoint Inhibitors in Hepatocellular Carcinoma: an Overview
    pharmaceuticals Review Immune Checkpoint Inhibitors in Hepatocellular Carcinoma: An Overview Diederick J. van Doorn 1 , Robert Bart Takkenberg 1 and Heinz-Josef Klümpen 2,* 1 Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; [email protected] (D.J.v.D.); [email protected] (R.B.T.) 2 Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands * Correspondence: [email protected]; Tel.: +31-20-566-5983 Abstract: Patients with hepatocellular carcinoma (HCC) face a common type of cancer, which is amongst the most deadly types of cancer worldwide. The therapeutic options range from curative resection or ablation to loco regional therapies in palliative setting and last but not least, systemic treatment. The latter group underwent major changes in the last decade and a half. Since the introduction of sorafenib in 2007, many other systemic treatments have been investigated. Most without success. It took more than ten years before lenvatinib could be added as alternative first-line treatment option. Just recently a new form of systemic treatment, immunotherapy, entered the field of therapeutic options in patients with HCC. Immune checkpoint inhibitors are becoming the new standard of care in patients with HCC. Several reviews reported on the latest phase 1/2 studies and discussed the higher response rates and better tolerability when compared to current standard of care therapies. This review will focus on elaborating the working mechanism of these checkpoint inhibitors, give an elaborate update of the therapeutic agents that are currently available or under research, and will give an overview of the latest trials, as well as ongoing and upcoming trials.
    [Show full text]
  • Follow-Up and Management of Checkpoint Inhibitor Related Toxicities in Cancer Patients
    Guideline Resource Unit [email protected] Follow-up and Management of Checkpoint Inhibitor Related Toxicities in Cancer Patients Effective Date: July, 2020 Clinical Practice Guideline SUPP-018 – Version 1 www.ahs.ca/guru Table of Contents Background............................................................................................................................................3 Guideline Question.................................................................................................................................4 Search Strategy......................................................................................................................................4 Target Population...................................................................................................................................4 Recommendations General Approaches to Toxicity Management…....................................................................................5 Dermatology Prevention...........................................................................................................................................5 Anticipation..........................................................................................................................................6 Detection.............................................................................................................................................6 Treatment and Monitoring...................................................................................................................7
    [Show full text]
  • Immune Checkpoint Inhibitors: a Strategy to Tackle Cancer?
    Immune checkpoint inhibitors: A strategy to tackle cancer? Commentary by Alessia Armezzani, PhD 02/07/2019 Our immune system is constantly on the lookout for pathogens and, once it encounters one, it mounts a strategic attack to fight it: it is the so-called immune response. To minimize the potential collateral damage to healthy cells and tissues though, this response needs to be tuned down.(1, 2) And that is precisely the role of inhibitory immune checkpoints, a plethora of molecules naturally expressed both on T cells and antigen-presenting cells that maintains self-tolerance and limits tissue damage by recognizing ligands expressed on self-tissues.(3) In the past 30 years, a broad range of immune checkpoint molecules have been identified, including cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1) whose discoveries have earned James P. Allison and Tasuku Honjo the Nobel Prize in medicine in 2018. Both proteins act as negative regulators of T cell activation, thereby preventing unwanted immune responses.(4, 5, 6, 7, 8) Immune checkpoints are also expressed on many tumor cells, allowing them to cleverly evade host immune response and divide uncontrollably.(9) Interestingly, several in vivo studies have demonstrated that antibodies directed against key immune checkpoints such as PD-1 and CTLA-4 inhibit their function, thereby allowing the elimination of certain tumor cells.(2, 10, 11, 12) Without With These findings have provided a rationale for targeting immunotherapy immunotherapy immune
    [Show full text]
  • Immunotherapy in Hodgkin Lymphoma: Present Status and Future Strategies
    cancers Review Immunotherapy in Hodgkin Lymphoma: Present Status and Future Strategies Theodoros P. Vassilakopoulos * , Chrysovalantou Chatzidimitriou, John V. Asimakopoulos, Maria Arapaki, Evangelos Tzoras, Maria K. Angelopoulou and Kostas Konstantopoulos Department of Haematology and Bone Marrow Transplantation, National and Kapodistrian University of Athens, Laikon General Hospital, 11527 Athens, Greece * Correspondence: [email protected]; Tel.: +30-213-2061702; Fax: +30-213-2061498 Received: 8 May 2019; Accepted: 13 June 2019; Published: 29 July 2019 Abstract: Although classical Hodgkin lymphoma (cHL) is usually curable, 20–30% of the patients experience treatment failure and most of them are typically treated with salvage chemotherapy and autologous stem cell transplantation (autoSCT). However, 45–55% of that subset further relapse or progress despite intensive treatment. At the advanced stage of the disease course, recently developed immunotherapeutic approaches have provided very promising results with prolonged remissions or disease stabilization in many patients. Brentuximab vedotin (BV) has been approved for patients with relapsed/refractory cHL (rr-cHL) who have failed autoSCT, as a consolidation after autoSCT in high-risk patients, as well as for patients who are ineligible for autoSCT or multiagent chemotherapy who have failed two treatment lines. However, except of the consolidation setting, 90–95% of the ≥ patients will progress and require further treatment. In this clinical setting, immune checkpoint inhibitors (CPIs) have produced impressive results. Both nivolumab and pembrolizumab have been approved for rr-cHL after autoSCT and BV failure, while pembrolizumab has also been licensed for transplant ineligible patients after BV failure. Other CPIs, sintilimab and tislelizumab, have been successfully tested in China, albeit in less heavily pretreated populations.
    [Show full text]
  • Review Article Immune Checkpoint Inhibitor Nephrotoxicity
    Review Article Immune Checkpoint Inhibitor Nephrotoxicity: Update 2020 Shruti Gupta ,1 Frank B. Cortazar,2,3 Leonardo V. Riella,1 and David E. Leaf1 Abstract Immune checkpoint inhibitors (ICPIs) have transformed the landscape of oncology, but are associated with a variety of autoimmune adverse events, including AKI. ICPI-associated AKI (ICPI-AKI) is emerging as an increasingly frequent cause of AKI in patients with cancer, and poses unique diagnostic and management challenges to clinicians who care for these patients. In this review, we describe the incidence and risk factors for ICPI-AKI, including proton pump inhibitor use, CKD, and combination immunotherapy. We discuss the limitations of the various definitions used for ICPI-AKI in prior studies, and propose a novel classification system (definite, probable, and possible ICPI-AKI) that recognizes the diagnostic uncertainty inherent in many cases. We discuss the key clinicopathologic features and treatment strategies for ICPI-AKI, including the role of kidney biopsy versus empirical treatment with steroids. We also explore the under-studied area of ICPI use in the setting of solid organ transplantation, where nephrologists and oncologists must balance the risk of rejection versus treating the underlying malignancy. Finally, we summarize existing data on the role of ICPI rechallenge after an episode of ICPI-AKI. KIDNEY360 1: 130–140, 2020. doi: https://doi.org/10.34067/KID.0000852019 Introduction therapy, and the potential for irreversible organ dam- Targeted immune therapies have transformed the age. Here we highlight ten burning questions on ICPI- landscape of oncology, with immune checkpoint inhib- AKI. We discuss our current understanding of the itors (ICPIs) at the forefront.
    [Show full text]