1
|
Grimaldi C, Ibraghimov A, Kiessling A, Rattel B, Ji C, Fuller CL, Brennan FR, Regenass-Lechner F, Shenton J, Price KD, Piché MS, Steeves MA, Prell R, Dudal S, Kronenberg S, Freebern W, Blanset D. Current nonclinical approaches for immune assessments of immuno-oncology biotherapeutics. Drug Discov Today 2023; 28:103440. [PMID: 36375739 DOI: 10.1016/j.drudis.2022.103440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/30/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Harnessing the immune system to kill tumors has been revolutionary and, as a result, has had an enormous benefit for patients in extending life and resulting in effective cures in some. However, activation of the immune system can come at the cost of undesirable adverse events such as cytokine release syndrome, immune-related adverse events, on-target/off-tumor toxicity, neurotoxicity and tumor lysis syndrome, which are safety risks that can be challenging to assess non-clinically. This article provides a review of the biology and mechanisms that can result in immune-mediated adverse effects and describes industry approaches using in vitro and in vivo models to aid in the nonclinical safety risk assessments for immune-oncology modalities. Challenges and limitations of knowledge and models are also discussed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Sherri Dudal
- Roche Pharmaceutical Research and Early Development, United States
| | - Sven Kronenberg
- Roche Pharmaceutical Research and Early Development, United States
| | | | - Diann Blanset
- Boehringer Ingelheim Pharmaceuticals, Inc., United States.
| |
Collapse
|
2
|
Chen T, Sun T, Bian Y, Pei Y, Feng F, Chi H, Li Y, Tang X, Sang S, Du C, Chen Y, Chen Y, Sun H. The Design and Optimization of Monomeric Multitarget Peptides for the Treatment of Multifactorial Diseases. J Med Chem 2022; 65:3685-3705. [DOI: 10.1021/acs.jmedchem.1c01456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Tingkai Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Tianyu Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Yaoyao Bian
- College of Acupuncture and Massage, College of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing 210023, People’s Republic of China
| | - Yuqiong Pei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People’s Republic of China
| | - Feng Feng
- Food and Pharmaceutical Research Institute, Jiangsu Food and Pharmaceuticals Science College, Huaian 223003, People’s Republic of China
| | - Heng Chi
- Food and Pharmaceutical Research Institute, Jiangsu Food and Pharmaceuticals Science College, Huaian 223003, People’s Republic of China
| | - Yuan Li
- Department of Pharmaceutical Engineering, Jiangsu Food and Pharmaceuticals Science College, Huaian 223005, People’s Republic of China
| | - Xu Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People’s Republic of China
| | - Shenghu Sang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People’s Republic of China
| | - Chenxi Du
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Ying Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People’s Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| |
Collapse
|
3
|
Zheng B, Zhang P, Wang H, Wang J, Liu ZH, Zhang D. Advances in Research on Bladder Cancer Targeting Peptides: a Review. Cell Biochem Biophys 2021; 79:711-718. [PMID: 34468956 PMCID: PMC8558283 DOI: 10.1007/s12013-021-01019-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2021] [Indexed: 12/04/2022]
Abstract
Bladder cancer (Bca) is the second most common malignant tumor of the genitourinary system in Chinese male population with high potential of recurrence and progression. The overall prognosis has not been improved significantly for the past 30 years due to the lack of early theranostic technique. Currently the early theranostic technique for bladder cancer is mainly through the intravesical approach, but the clinical outcomes are poor due to the limited tumor-targeting efficiency. Therefore, the targeting peptides for bladder cancer provide possibility to advance intravesical theranostic technique. However, no systematic review has covered the wide use of the targeting peptides for intravesical theranostic techniques in bladder cancer. Herein, a summary of original researches introduces all aspects of the targeting peptides for bladder cancer, including the peptide screening, the targeting mechanism and its preclinical application.
Collapse
Affiliation(s)
- Bin Zheng
- Zhejiang Chinese Medical University, 310053, HangZhou, China
- Zhejiang Provincial People's Hospital, Hangzhou Medical College, 310014, Hangzhou, China
| | - Pu Zhang
- Zhejiang Provincial People's Hospital, Hangzhou Medical College, 310014, Hangzhou, China
| | - Heng Wang
- Zhejiang Provincial People's Hospital, Hangzhou Medical College, 310014, Hangzhou, China
| | - Jinxue Wang
- Handan Central hospital, 056001, Handan, China
| | - Zheng Hong Liu
- Zhejiang Chinese Medical University, 310053, HangZhou, China
| | - DaHong Zhang
- Zhejiang Chinese Medical University, 310053, HangZhou, China.
| |
Collapse
|
4
|
Harnessing the combined potential of cancer immunotherapy and nanomedicine: A new paradigm in cancer treatment. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 40:102492. [PMID: 34775062 DOI: 10.1016/j.nano.2021.102492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 10/16/2021] [Accepted: 10/29/2021] [Indexed: 11/21/2022]
Abstract
Cancer immunotherapy has recently emerged as a rising star due to its ability to activate patients' immune systems to fight tumors and prevent relapse. Conversely, the interest in cancer nanomedicine has seemingly waned due to its lackluster clinical translation. Despite being hailed as a game-changer in oncology, cancer immunotherapy still faces numerous challenges. Combining both entities together has thus been one among several solutions proposed to circumvent these challenges. This solution has since gained traction and has also led to a renaissance of cancer nanomedicine. While most combinations are currently experimental at best, some have progressed on to clinical trials. This review thus seeks to examine the advantages and disadvantages of integrating both modalities as a cancer treatment. The opportunities, challenges and future directions of this emerging field will also be explored with the hope that such a combination will lead to a paradigm shift in cancer treatments.
Collapse
|
5
|
Correa S, Grosskopf AK, Lopez Hernandez H, Chan D, Yu AC, Stapleton LM, Appel EA. Translational Applications of Hydrogels. Chem Rev 2021; 121:11385-11457. [PMID: 33938724 PMCID: PMC8461619 DOI: 10.1021/acs.chemrev.0c01177] [Citation(s) in RCA: 395] [Impact Index Per Article: 98.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Indexed: 12/17/2022]
Abstract
Advances in hydrogel technology have unlocked unique and valuable capabilities that are being applied to a diverse set of translational applications. Hydrogels perform functions relevant to a range of biomedical purposes-they can deliver drugs or cells, regenerate hard and soft tissues, adhere to wet tissues, prevent bleeding, provide contrast during imaging, protect tissues or organs during radiotherapy, and improve the biocompatibility of medical implants. These capabilities make hydrogels useful for many distinct and pressing diseases and medical conditions and even for less conventional areas such as environmental engineering. In this review, we cover the major capabilities of hydrogels, with a focus on the novel benefits of injectable hydrogels, and how they relate to translational applications in medicine and the environment. We pay close attention to how the development of contemporary hydrogels requires extensive interdisciplinary collaboration to accomplish highly specific and complex biological tasks that range from cancer immunotherapy to tissue engineering to vaccination. We complement our discussion of preclinical and clinical development of hydrogels with mechanical design considerations needed for scaling injectable hydrogel technologies for clinical application. We anticipate that readers will gain a more complete picture of the expansive possibilities for hydrogels to make practical and impactful differences across numerous fields and biomedical applications.
Collapse
Affiliation(s)
- Santiago Correa
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Abigail K. Grosskopf
- Chemical
Engineering, Stanford University, Stanford, California 94305, United States
| | - Hector Lopez Hernandez
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Doreen Chan
- Chemistry, Stanford University, Stanford, California 94305, United States
| | - Anthony C. Yu
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
| | | | - Eric A. Appel
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
- Bioengineering, Stanford University, Stanford, California 94305, United States
- Pediatric
Endocrinology, Stanford University School
of Medicine, Stanford, California 94305, United States
- ChEM-H Institute, Stanford
University, Stanford, California 94305, United States
- Woods
Institute for the Environment, Stanford
University, Stanford, California 94305, United States
| |
Collapse
|
6
|
Peptide-based and small molecule PD-1 and PD-L1 pharmacological modulators in the treatment of cancer. Pharmacol Ther 2021; 227:107870. [PMID: 33895183 DOI: 10.1016/j.pharmthera.2021.107870] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/09/2021] [Accepted: 04/20/2021] [Indexed: 12/15/2022]
Abstract
Cancer immunotherapy is an option to enhance physiological defence mechanism to fight cancer, where natural substances (e.g., antigen/antibody) or small synthetic molecule can be utilized to improve and restore the immune system to stop or slacken the development of malignant cells, stop metastasis and/or help the immune response with synthetic monoclonal antibodies (mAbs) and tumour-agnostic therapy to eliminate cancer cells. Interaction between the programmed cell death ligand 1 (PD-L1) and its receptor (programmed cell death protein 1, PD-1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA4) linked signalling pathways have been identified as perilous towards the body's immune mechanism in regulating the progression of cancer. It is known that certain cancers use these pathways to evade the body's defence mechanism. The immune system is capable of responding to cancer by stalling these trails with specific synthetic antibodies or immune checkpoint inhibitors, which can ultimately either stop or slow cancer cell development. Recent findings and data suggested that using such inhibitors invigorated a new approach to cancer treatment. These inhibitors usually activate the immune system to identify and eliminate cancer cells rather than attacking tumour cells directly. PD-1/PD-L1 inhibitors have already been substantiated for their efficacy in over twenty variations of cancer through different clinical trials. Studies on molecular interaction with existing PD-1/PD-L1 inhibitors that are mainly dominated by antibodies are constantly generating new ideas to develop novel inhibitors. This review has summarised information on reported and/or patented small molecules and peptides for their ability to interact with the PD-1/PD-L1 as a potential anticancer strategy.
Collapse
|
7
|
Abstract
Tumor-homing peptides are widely used for improving tumor selectivity of anticancer drugs and imaging agents. The goal is to increase tumor uptake and reduce accumulation at nontarget sites. Here, we describe current approaches for tumor-homing peptide identification and validation, and provide comprehensive overview of classes of tumor-homing peptides undergoing preclinical and clinical development. We focus on unique mechanistic features and applications of a recently discovered class of tumor-homing peptides, tumor-penetrating C-end Rule (CendR) peptides, that can be used for tissue penetrative targeting of extravascular tumor tissue. Finally, we discuss unanswered questions and future directions in the field of development of peptide-guided smart drugs and imaging agents.
Collapse
|
8
|
Chames P, Wurch T. [Bispecific antibodies, novel therapeutic candidates harnessing the immune system]. Med Sci (Paris) 2020; 35:1072-1082. [PMID: 31903920 DOI: 10.1051/medsci/2019242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Over the past ten years, an increased knowledge of tumor biology and immunology allowed the design and development of novel therapeutic antibody and protein scaffold formats, where bispecific antibodies (Abs) play a major role. The latter molecules can (1) bring novel pharmacological properties through the co-engagement of two targets, (2) increase the safety profile as compared to a combination of two antibodies thanks to a targeted relocation to the tumor and (3) reduce development and manufacturing costs associated with single drug product. This review analyzes the different bispecific antibodies and scaffolds described in the field of immuno-oncology, their structure and major pharmacological and physico-chemical properties.
Collapse
Affiliation(s)
- Patrick Chames
- Aix Marseille Univ, CNRS, Inserm, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Thierry Wurch
- Centre d'Innovation Thérapeutique en Oncologie - Servier, F78290 Croissy-sur-Seine, France
| |
Collapse
|
9
|
Leufven E, Bruserud Ø. Immunosuppression and Immunotargeted Therapy in Acute Myeloid Leukemia - The Potential Use of Checkpoint Inhibitors in Combination with Other Treatments. Curr Med Chem 2019; 26:5244-5261. [PMID: 30907305 DOI: 10.2174/0929867326666190325095853] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/12/2019] [Accepted: 03/12/2019] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Immunotherapy by using checkpoint inhibitors is now tried in the treatment of several malignancies, including Acute Myeloid Leukemia (AML). The treatment is tried both as monotherapy and as a part of combined therapy. METHODS Relevant publications were identified through literature searches in the PubMed database. We searched for (i) original articles describing the results from clinical studies of checkpoint inhibition; (ii) published articles describing the immunocompromised status of AML patients; and (iii) published studies of antileukemic immune reactivity and immunotherapy in AML. RESULTS Studies of monotherapy suggest that checkpoint inhibition has a modest antileukemic effect and complete hematological remissions are uncommon, whereas combination with conventional chemotherapy increases the antileukemic efficiency with acceptable toxicity. The experience with a combination of different checkpoint inhibitors is limited. Thalidomide derivatives are referred to as immunomodulatory drugs and seem to reverse leukemia-induced immunosuppression, but in addition, they have direct inhibitory effects on the AML cells. The combination of checkpoint targeting and thalidomide derivatives thus represents a strategy for dual immunotargeting together with a direct antileukemic effect. CONCLUSION Checkpoint inhibitors are now tried in AML. Experimental studies suggest that these inhibitors should be combined with immunomodulatory agents (i.e. thalidomide derivatives) and/or new targeted or conventional antileukemic treatment. Such combinations would allow dual immunotargeting (checkpoint inhibitor, immunomodulatory agents) together with a double/triple direct targeting of the leukemic cells.
Collapse
Affiliation(s)
- Eva Leufven
- Department of Clinical Science, University of Bergen, Jonas Lies vei 87, N-5020 Bergen, Norway
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, Jonas Lies vei 87, N-5020 Bergen, Norway.,Section for Hematology, Department of Medicine, Haukeland University Hospital, N-5021, Bergen, Norway
| |
Collapse
|
10
|
Richards DM, Marschall V, Billian-Frey K, Heinonen K, Merz C, Redondo Müller M, Sefrin JP, Schröder M, Sykora J, Fricke H, Hill O, Gieffers C, Thiemann M. HERA-GITRL activates T cells and promotes anti-tumor efficacy independent of FcγR-binding functionality. J Immunother Cancer 2019; 7:191. [PMID: 31324216 PMCID: PMC6642547 DOI: 10.1186/s40425-019-0671-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/08/2019] [Indexed: 11/28/2022] Open
Abstract
Background Glucocorticoid-induced TNFR-related protein (TNFRSF18, GITR, CD357), expressed by T cells, and its ligand (TNFSF18, GITRL), expressed by myeloid populations, provide co-stimulatory signals that boost T cell activity. Due to the important role that GITR plays in regulating immune functions, agonistic stimulation of GITR is a promising therapeutic concept. Multiple strategies to induce GITR signaling have been investigated. The limited clinical efficacy of antibody-based GITR agonists results from structural and functional characteristics of antibodies that are unsuitable for stimulating the well-defined trimeric members of the TNFRSF. Methods To overcome limitations of antibody-based TNFRSF agonists, we have developed HERA-GITRL, a fully human hexavalent TNF receptor agonist (HERA) targeting GITR and mimicking the natural signaling concept. HERA-GITRL is composed of a trivalent but single-chain GITRL-receptor-binding-domain (scGITRL-RBD) unit fused to an IgG1 derived silenced Fc-domain serving as dimerization scaffold. A specific mouse surrogate, mmHERA-GITRL, was also generated to examine in vivo activity in respective mouse tumor models. Results For functional characterization of HERA-GITRL in vitro, human immune cells were isolated from healthy-donor blood and stimulated with anti-CD3 antibody in the presence of HERA-GITRL. Consistently, HERA-GITRL increased the activity of T cells, including proliferation and differentiation, even in the presence of regulatory T cells. In line with these findings, mmHERA-GITRL enhanced antigen-specific clonal expansion of both CD4+ (OT-II) and CD8+ (OT-I) T cells in vivo while having no effect on non-specific T cells. In addition, mmHERA-GITRL showed single-agent anti-tumor activity in two subcutaneous syngeneic colon cancer models (CT26wt and MC38-CEA). Importantly, this activity is independent of its FcγR-binding functionality, as both mmHERA-GITRL with a functional Fc- and a silenced Fc-domain showed similar tumor growth inhibition. Finally, in a direct in vitro comparison to a bivalent clinical benchmark anti-GITR antibody and a trivalent GITRL, only the hexavalent HERA-GITRL showed full biological activity independent of additional crosslinking. Conclusion In this manuscript, we describe the development of HERA-GITRL, a true GITR agonist with a clearly defined mechanism of action. By clustering six receptor chains in a spatially well-defined manner, HERA-GITRL induces potent agonistic activity without being dependent on additional FcγR-mediated crosslinking. Electronic supplementary material The online version of this article (10.1186/s40425-019-0671-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- David M Richards
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | | | - Katharina Billian-Frey
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | - Karl Heinonen
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | - Christian Merz
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | | | - Julian P Sefrin
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | - Matthias Schröder
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | - Jaromir Sykora
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | | | - Oliver Hill
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | - Christian Gieffers
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany
| | - Meinolf Thiemann
- Research and Development, Apogenix AG, Im Neuenheimer Feld 584, 69120, Heidelberg, Germany.
| |
Collapse
|
11
|
Thiemann M, Richards DM, Heinonen K, Kluge M, Marschall V, Merz C, Redondo Müller M, Schnyder T, Sefrin JP, Sykora J, Fricke H, Gieffers C, Hill O. A Single-Chain-Based Hexavalent CD27 Agonist Enhances T Cell Activation and Induces Anti-Tumor Immunity. Front Oncol 2018; 8:387. [PMID: 30298117 PMCID: PMC6160747 DOI: 10.3389/fonc.2018.00387] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 08/29/2018] [Indexed: 01/05/2023] Open
Abstract
Tumor necrosis factor receptor superfamily member 7 (TNFRSF7, CD27), expressed primarily by T cells, and its ligand CD27L (TNFSF7, CD70) provide co-stimulatory signals that boost T cell activation, differentiation, and survival. Agonistic stimulation of CD27 is therefore a promising therapeutic concept in immuno-oncology intended to boost and sustain T cell driven anti-tumor responses. Endogenous TNFSF/TNFRSF-based signal transmission is a structurally well-defined event that takes place during cell-to-cell-based contacts. It is well-established that the trimeric-trivalent TNFSF-receptor binding domain (TNFSF-RBD) exposed by the conducting cell and the resulting multi-trimer-based receptor clustering on the receiving cell are essential for agonistic signaling. Therefore, we have developed HERA-CD27L, a novel hexavalent TNF receptor agonist (HERA) targeting CD27 and mimicking the natural signaling concept. HERA-CD27L is composed of a trivalent but single-chain CD27L-receptor-binding-domain (scCD27L-RBD) fused to an IgG1 derived silenced Fc-domain serving as dimerization scaffold. The hexavalent agonist significantly boosted antigen-specific T cell responses while having no effect on non-specific T cells and was superior over stabilized recombinant trivalent CD27L. In addition, HERA-CD27L demonstrated potent single-agent anti-tumor efficacy in two different syngeneic tumor models, MC38-CEA and CT26wt. Furthermore, the combination of HERA-CD27L and an anti-PD-1 antibody showed additive anti-tumor effects highlighting the importance of both T cell activation and checkpoint inhibition in anti-tumor immunity. In this manuscript, we describe the development of HERA-CD27L, a true CD27 agonist with a clearly defined forward-signaling mechanism of action.
Collapse
|
12
|
Konstorum A, Vella AT, Adler AJ, Laubenbacher RC. Addressing current challenges in cancer immunotherapy with mathematical and computational modelling. J R Soc Interface 2018; 14:rsif.2017.0150. [PMID: 28659410 DOI: 10.1098/rsif.2017.0150] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/31/2017] [Indexed: 02/06/2023] Open
Abstract
The goal of cancer immunotherapy is to boost a patient's immune response to a tumour. Yet, the design of an effective immunotherapy is complicated by various factors, including a potentially immunosuppressive tumour microenvironment, immune-modulating effects of conventional treatments and therapy-related toxicities. These complexities can be incorporated into mathematical and computational models of cancer immunotherapy that can then be used to aid in rational therapy design. In this review, we survey modelling approaches under the umbrella of the major challenges facing immunotherapy development, which encompass tumour classification, optimal treatment scheduling and combination therapy design. Although overlapping, each challenge has presented unique opportunities for modellers to make contributions using analytical and numerical analysis of model outcomes, as well as optimization algorithms. We discuss several examples of models that have grown in complexity as more biological information has become available, showcasing how model development is a dynamic process interlinked with the rapid advances in tumour-immune biology. We conclude the review with recommendations for modellers both with respect to methodology and biological direction that might help keep modellers at the forefront of cancer immunotherapy development.
Collapse
Affiliation(s)
- Anna Konstorum
- Center for Quantitative Medicine, UConn Health, Farmington, CT, USA
| | | | - Adam J Adler
- Department of Immunology, UConn Health, Farmington, CT, USA
| | - Reinhard C Laubenbacher
- Center for Quantitative Medicine, UConn Health, Farmington, CT, USA .,Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| |
Collapse
|
13
|
Seek & Destroy, use of targeting peptides for cancer detection and drug delivery. Bioorg Med Chem 2017; 26:2797-2806. [PMID: 28893601 DOI: 10.1016/j.bmc.2017.08.052] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/14/2017] [Accepted: 08/30/2017] [Indexed: 12/21/2022]
Abstract
Accounting for 16 million new cases and 9 million deaths annually, cancer leaves a great number of patients helpless. It is a complex disease and still a major challenge for the scientific and medical communities. The efficacy of conventional chemotherapies is often poor and patients suffer from off-target effects. Each neoplasm exhibits molecular signatures - sometimes in a patient specific manner - that may completely differ from the organ of origin, may be expressed in markedly higher amounts and/or in different location compared to the normal tissue. Although adding layers of complexity in the understanding of cancer biology, this cancer-specific signature provides an opportunity to develop targeting agents for early detection, diagnosis, and therapeutics. Chimeric antibodies, recombinant proteins or synthetic polypeptides have emerged as excellent candidates for specific homing to peripheral and central nervous system cancers. Specifically, peptide ligands benefit from their small size, easy and affordable production, high specificity, and remarkable flexibility regarding their sequence and conjugation possibilities. Coupled to imaging agents, chemotherapies and/or nanocarriers they have shown to increase the on-site delivery, thus allowing better tumor mass contouring in imaging and increased efficacy of the chemotherapies associated with reduced adverse effects. Therefore, some of the peptides alone or in combination have been tested in clinical trials to treat patients. Peptides have been well-tolerated and shown absence of toxicity. This review aims to offer a view on tumor targeting peptides that are either derived from natural peptide ligands or identified using phage display screening. We also include examples of peptides targeting the high-grade malignant tumors of the central nervous system as an example of the complex therapeutic management due to the tumor's location. Peptide vaccines are outside of the scope of this review.
Collapse
|
14
|
Cabo M, Offringa R, Zitvogel L, Kroemer G, Muntasell A, Galluzzi L. Trial Watch: Immunostimulatory monoclonal antibodies for oncological indications. Oncoimmunology 2017; 6:e1371896. [PMID: 29209572 PMCID: PMC5706611 DOI: 10.1080/2162402x.2017.1371896] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 08/21/2017] [Indexed: 12/14/2022] Open
Abstract
The goal of cancer immunotherapy is to establish new or boost pre-existing anticancer immune responses that eradicate malignant cells while generating immunological memory to prevent disease relapse. Over the past few years, immunomodulatory monoclonal antibodies (mAbs) that block co-inhibitory receptors on immune effectors cells - such as cytotoxic T lymphocyte-associated protein 4 (CTLA4), programmed cell death 1 (PDCD1, best known as PD-1) - or their ligands - such as CD274 (best known as PD-L1) - have proven very successful in this sense. As a consequence, many of such immune checkpoint blockers (ICBs) have already entered the clinical practice for various oncological indications. Considerable attention is currently being attracted by a second group of immunomodulatory mAbs, which are conceived to activate co-stimulatory receptors on immune effector cells. Here, we discuss the mechanisms of action of these immunostimulatory mAbs and summarize recent progress in their preclinical and clinical development.
Collapse
Affiliation(s)
- Mariona Cabo
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Rienk Offringa
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
- DKFZ-Bayer Joint Immunotherapeutics Laboratory, German Cancer Research Center, Heidelberg, Germany
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, U1015, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, France
- Université Pierre et Marie Curie/Paris VI, Paris
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
- Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP; Paris, France
| | - Aura Muntasell
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, France
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
15
|
Bourke JM, O'Sullivan M, Khattak MA. Management of adverse events related to new cancer immunotherapy (immune checkpoint inhibitors). Med J Aust 2017; 205:418-424. [PMID: 27809739 DOI: 10.5694/mja16.00586] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 07/20/2016] [Indexed: 12/19/2022]
Abstract
New immunotherapies have significantly improved survival in certain advanced cancers in recent years, particularly metastatic melanoma and lung cancer. The most effective of these therapies are the immune checkpoint inhibitors (ICIs) such as ipilimumab, nivolumab and pembrolizumab. The use of ICIs will continue to increase in the coming years as evidence of their benefit in a range of other cancers builds. ICIs are associated with novel immune-related adverse events (irAEs), which can involve a wide range of organs. The most common irAEs involve the skin (rash, pruritus), gastrointestinal tract (diarrhoea, colitis) and endocrine system (thyroid, pituitary). While severity is generally mild, life-threatening complications can occur if not recognised and treated promptly. Due to the diverse manifestations of irAEs, patients may present to doctors who are not familiar with these drugs, which creates the potential for delays in management. Management of irAEs depends on severity and the organ affected. Systemic steroids are often required and ICI therapy may be withheld or discontinued. Additional immunosuppressive medications may be necessary in steroid-refractory cases. This review provides an overview of the potential toxicities and their management for general clinicians. Broader awareness of these issues among medical professionals will hopefully reduce unnecessary delays in diagnosis and treatment. Patient and carer education regarding irAEs is extremely important; patients and carers should be advised to seek urgent medical attention if required.
Collapse
|
16
|
In vitro assays supporting the safety assessment of immunomodulatory monoclonal antibodies. Toxicol In Vitro 2017; 45:296-308. [PMID: 28263892 DOI: 10.1016/j.tiv.2017.02.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 02/08/2017] [Accepted: 02/28/2017] [Indexed: 01/11/2023]
Abstract
Many monoclonal antibodies (mAbs) licensed for human use or in clinical development for cancer and autoimmune disease directly interact with the immune system. These immunomodulatory mAbs have an inherent risk of adverse immune-mediated drug reactions, including infusion reactions, cytokine storms, immunosuppression and autoimmunity. A thorough understanding of the potential for immunotoxicity of a mAb is required to support administration to humans. This review will highlight the key role of in vitro assays in defining the immunopharmacology, immunotoxicity and immunogenicity of mAbs. A wide range of in vitro tests with multiple formats of different complexity can be utilized to characterize i) the antibody-binding domains of the mAb, such as on-target binding and downstream pharmacological effects (e.g. immunosuppression, immune activation, cytokine release) in both humans and animal species used for toxicology studies and off-target binding; ii) Fc-dependent effects such as Fc-mediated cellular activation (e.g. of leukocytes, platelets) and cytokine release, complement activation; and iii) product-related factors (sequence, physical-chemical properties and impurities) that can impact both pharmacological activity and immunogenicity potential of a mAb. These assays can be crucial to the selection of mAbs with an optimum balance of safety and efficacy, in defining whether a mAb is a high risk molecule, and together with animal data, can inform human safe starting doses and escalation schemes.
Collapse
|
17
|
Brennan FR, Kiessling A. Translational immunotoxicology of immunomodulatory monoclonal antibodies. DRUG DISCOVERY TODAY. TECHNOLOGIES 2016; 21-22:85-93. [PMID: 27978992 DOI: 10.1016/j.ddtec.2016.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/17/2016] [Accepted: 08/18/2016] [Indexed: 12/28/2022]
Abstract
While immunomodulatory monoclonal antibodies (mAbs) have a wide therapeutic potential, exaggerated immunopharmacology may drive both acute and delayed immunotoxicity. The existing tools for immunotoxicity assessment do not accurately predict the full range of immunotoxicities observed in humans. New and optimized models, assays, endpoints and biomarkers in animals and humans are required to safeguard patients and allow them access to these often transformational therapies.
Collapse
|
18
|
Davis RJ, Ferris RL, Schmitt NC. Costimulatory and coinhibitory immune checkpoint receptors in head and neck cancer: unleashing immune responses through therapeutic combinations. CANCERS OF THE HEAD & NECK 2016; 1:12. [PMID: 31093342 PMCID: PMC6460794 DOI: 10.1186/s41199-016-0013-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/01/2016] [Indexed: 06/09/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) represents a model of escape from anti-tumor immunity. The high frequency of p53 tumor suppressor loss in HNSCC leads to genomic instability and immune stimulation through the generation of neoantigens. However, the aggressive nature of HNSCC tumors and significant rates of resistance to conventional therapies highlights the ability of HNSCC to evade this immune response. Advances in understanding the role of co-stimulatory and immune checkpoint receptors in HNSCC-mediated immunosuppression lay the foundation for development of novel therapeutic approaches. This article provides an overview of these co-stimulatory and immune checkpoint pathways, as well as a review of preclinical and clinical evidence supporting the modulation of these pathways in HNSCC. Finally, the synergistic potential of combining these approaches is discussed, along with an update of current clinical trials evaluating combinations of immune-based therapies in HNSCC patients.
Collapse
Affiliation(s)
- Ruth J. Davis
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, 10 Center Drive, Room 5B-39, Bethesda, MD 20892 USA
| | - Robert L. Ferris
- Department of Otolaryngology, Hillman Cancer Center Research Pavilion, University of Pittsburgh, 5117 Centre Avenue, Room 2.26b, Pittsburgh, PA 15213-1863 USA
- Department of Immunology, Hillman Cancer Center Research Pavilion, University of Pittsburgh, 5117 Centre Avenue, Room 2.26b, Pittsburgh, PA 15213-1863 USA
- Cancer Immunology Programm, Hillman Cancer Center Research Pavilion, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Room 2.26b, Pittsburgh, PA 15213-1863 USA
| | - Nicole C. Schmitt
- Tumor Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, 10 Center Drive, Room 5B-39, Bethesda, MD 20892 USA
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, 6420 Rockledge Drive, Suite 4920, Bethesda, MD 20817 USA
| |
Collapse
|