scholarly journals Challenges and Recent Advances in NK Cell-Targeted Immunotherapies in Solid Tumors

2021 ◽  
Vol 23 (1) ◽  
pp. 164
Author(s):  
Guang-Yu Lian ◽  
Thomas Shiu-Kwong Mak ◽  
Xue-Qing Yu ◽  
Hui-Yao Lan

Natural killer (NK) cell is a powerful malignant cells killer, providing rapid immune responses via direct cytotoxicity without the need of antigen processing and presentation. It plays an essential role in preventing early tumor, metastasis and minimal residual disease. Although adoptive NK therapies achieved great success in clinical trials against hematologic malignancies, their accumulation, activation, cytotoxic and immunoregulatory functions are severely impaired in the immunosuppressive microenvironment of solid tumors. Now with better understandings of the tumor evasive mechanisms from NK-mediated immunosurveillance, immunotherapies targeting the key molecules for NK cell dysfunction and exhaustion have been developed and tested in both preclinical and clinical studies. In this review, we introduce the challenges that NK cells encountered in solid tumor microenvironment (TME) and the therapeutic approaches to overcome these limitations, followed by an outline of the recent preclinical advances and the latest clinical outcomes of NK-based immunotherapies, as well as promising strategies to optimize current NK-targeted immunotherapies for solid tumors.

2021 ◽  
Vol 12 ◽  
Author(s):  
Sarah I. M. Sutherland ◽  
Xinsheng Ju ◽  
L. G. Horvath ◽  
Georgina J. Clark

Tumors evade the immune system though a myriad of mechanisms. Using checkpoint inhibitors to help reprime T cells to recognize tumor has had great success in malignancies including melanoma, lung, and renal cell carcinoma. Many tumors including prostate cancer are resistant to such treatment. However, Sipuleucel-T, a dendritic cell (DC) based immunotherapy, improved overall survival (OS) in prostate cancer. Despite this initial success, further DC vaccines have failed to progress and there has been limited uptake of Sipuleucel-T in the clinic. We know in prostate cancer (PCa) that both the adaptive and the innate arms of the immune system contribute to the immunosuppressive environment. This is at least in part due to dysfunction of DC that play a crucial role in the initiation of an immune response. We also know that there is a paucity of DC in PCa, and that those there are immature, creating a tolerogenic environment. These attributes make PCa a good candidate for a DC based immunotherapy. Ultimately, the knowledge gained by much research into antigen processing and presentation needs to translate from bench to bedside. In this review we will analyze why newer vaccine strategies using monocyte derived DC (MoDC) have failed to deliver clinical benefit, particularly in PCa, and highlight the emerging antigen loading and presentation technologies such as nanoparticles, antibody-antigen conjugates and virus co-delivery systems that can be used to improve efficacy. Lastly, we will assess combination strategies that can help overcome the immunosuppressive microenvironment of PCa.


Author(s):  
Shelagh Verco ◽  
Holly Maulhardt ◽  
Michael Baltezor ◽  
Emily Williams ◽  
Marc Iacobucci ◽  
...  

Abstract This report describes local administration of submicron particle paclitaxel (SPP) (NanoPac®: ~ 800-nm-sized particles with high relative surface area with each particle containing ~ 2 billion molecules of paclitaxel) in preclinical models and clinical trials evaluating treatment of carcinomas. Paclitaxel is active in the treatment of epithelial solid tumors including ovarian, peritoneal, pancreatic, breast, esophageal, prostate, and non-small cell lung cancer. SPP has been delivered directly to solid tumors, where the particles are retained and continuously release the drug, exposing primary tumors to high, therapeutic levels of paclitaxel for several weeks. As a result, tumor cell death shifts from primarily apoptosis to both apoptosis and necroptosis. Direct local tumoricidal effects of paclitaxel, as well as stimulation of innate and adaptive immune responses, contribute to antineoplastic effects. Local administration of SPP may facilitate tumor response to systemically administered chemotherapy, targeted therapy, or immunotherapy without contributing to systemic toxicity. Results of preclinical and clinical investigations described here suggest that local administration of SPP achieves clinical benefit with negligible toxicity and may complement standard treatments for metastatic disease. Graphical abstract


2021 ◽  
Vol 14 ◽  
Author(s):  
Wissam Zam ◽  
Amany Assaad

Background: Cancer is one of the leading causes of death worldwide. Chemotherapy, radiation therapy, and stem cell transplantation were the main cancer treatment approaches for several years but due to their limited effectiveness, there was a constant search for new therapeutic approaches. Cancer immunotherapy that utilizes and enhances the normal capacity of the patient's immune system was used to fight against cancer. Genetically engineered T-cells that express chimeric antigen receptors (CARs) showed remarkable anti-tumor activity against hematologic malignancies and is now being investigated in a variety of solid tumors. The use of this therapy in the last few years has been successful, achieving a great success in improving the quality of life and prolonging the survival time of patients with a reduction in remission rates. However, many challenges still need to be resolved in order for this technology to gain widespread adoption. Objective: This review summarizes various experimental approaches towards the use of CAR T-cells in hematologic malignancies and solid tumors. Conclusion: Finally, we address the challenges posed by CAR T-cells and discuss strategies for improving the performance of these T cells in fighting cancers.


Cancers ◽  
2019 ◽  
Vol 11 (7) ◽  
pp. 1040 ◽  
Author(s):  
Ziqing Chen ◽  
Ying Yang ◽  
Lisa L. Liu ◽  
Andreas Lundqvist

The immune system plays a crucial role to prevent local growth and dissemination of cancer. Therapies based on activating the immune system can result in beneficial responses in patients with metastatic disease. Treatment with antibodies targeting the immunological checkpoint axis PD-1 / PD-L1 can result in the induction of anti-tumor T cell activation leading to meaningful long-lasting clinical responses. Still, many patients acquire resistance or develop dose-limiting toxicities to these therapies. Analysis of tumors from patients who progress on anti-PD-1 treatment reveal defective interferon-signaling and antigen presentation, resulting in immune escape from T cell-mediated attack. Natural killer (NK) cells are innate lymphocytes that can kill tumor cells without prior sensitization to antigens and can be activated to kill tumor cells that have an impaired antigen processing and presentation machinery. Thus, NK cells may serve as useful effectors against tumor cells that have become resistant to classical immune checkpoint therapy. Various approaches to activate NK cells are being increasingly explored in clinical trials against cancer. While clinical benefit has been demonstrated in patients with acute myeloid leukemia receiving haploidentical NK cells, responses in patients with solid tumors are so far less encouraging. Several hurdles need to be overcome to provide meaningful clinical responses in patients with solid tumors. Here we review the recent developments to augment NK cell responses against solid tumors with regards to cytokine therapy, adoptive infusion of NK cells, NK cell engagers, and NK cell immune checkpoints.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1409-1409
Author(s):  
Sikander Ailawadhi ◽  
Paul Foster ◽  
Sarah Ryba ◽  
Michael Scharville ◽  
Assem El-Baghdady

Abstract Background: PD-(L)1 pathway inhibitors have demonstrated durable clinical benefit in both solid tumors and hematologic malignancies; however, as less than 1/3 of patients respond to anti-PD-(L)1 monotherapy, combination therapies may be needed for improved response rates and survival. T cell Immunoglobulin and ITIM domain (TIGIT) is expressed on activated T cell and natural killer (NK) cell subsets; TIGIT-CD155 binding suppresses T and NK cell function and inhibits the antitumor immune response. AB308, an Fc-enabled anti-TIGIT humanized IgG1 monoclonal antibody (mAb), has reversed TIGIT-CD155-mediated T cell inhibition in preclinical models. ARC-12 will assess whether TIGIT pathway blockade by AB308 can augment zimberelimab (zim; anti-PD-1 mAb) activity in patients with solid and hematologic malignancies. Study Design and Methods: ARC-12 is a phase 1/1b, first-in-human, open-label, dose-escalation study of AB308 + zim in patients with advanced solid tumors or hematologic malignancies. Dose expansion cohorts include relapsed/refractory (R/R) diffuse large B-cell lymphoma (DLBCL) and multiple myeloma (MM). For the dose-escalation, eligible adults have solid tumors for which no standard-of-care therapy exists or have pathologically-confirmed R/R non-Hodgkin lymphoma (NHL) and have not received or are ineligible for allogeneic stem cell transplant or adoptive cell transfer; ≤5 lines of prior systemic therapy; ≥1 measurable lesion per RECIST (solid tumors) or Lugano Classification (NHL) criteria; and ECOG performance score of 0-1. As shown in the figure, patients will receive AB308 + zim intravenously (IV) once every 3 weeks (Q3W; Part A) or once every 4 weeks (Q4W; Part B). In Part A, the first 6 patients of Cohort 1 will follow 3+3+6 rules, allowing for AB308 dose adjustment if dose-limiting toxicities (DLTs) warrant; all other dose-escalation cohorts will use 3+3 rules. After the Cohort 2 DLT period has been cleared, Part B enrollment will begin in parallel with Part A to determine the recommended dose for expansion (RDE) for AB308 + zim for both the Q3W (Part A) and Q4W (Part B) schedules. Each part can independently proceed to dose-expansion once the RDE is determined for that dose schedule. In the dose-expansion, patients will be enrolled into disease-specific cohorts for non-small cell lung cancer (NSCLC), melanoma, gastrointestinal cancer, cervical cancer, and hematologic malignancies (DLBCL or MM). AB308 + zim may be dosed on either a Q3W or Q4W schedule; all patients in each expansion cohort will receive the same dose schedule. For both the dose-escalation and dose-expansion, the primary endpoints assess safety and tolerability of AB308 + zim; secondary endpoints include pharmacokinetics (PK), immunogenicity, and objective response rate for AB308 + zim. Additional secondary endpoints in the dose-expansion are disease control rate and duration of response. Safety and efficacy data will be analyzed using summary statistics and Kaplan Meier estimates as appropriate; PK parameters will be estimated using noncompartmental methods. ARC-12 is actively recruiting at sites in the United States (NCT04772989). Figure 1 Figure 1. Disclosures Ailawadhi: Pharmacyclics: Consultancy, Research Funding; Takeda: Consultancy; Sanofi: Consultancy; Amgen: Consultancy, Research Funding; Ascentage: Research Funding; Genentech: Consultancy; AbbVie: Consultancy; BMS: Consultancy, Research Funding; Xencor: Research Funding; Janssen: Consultancy, Research Funding; Medimmune: Research Funding; Cellectar: Research Funding; Karyopharm: Consultancy; Beigene: Consultancy; GSK: Consultancy, Research Funding. Foster: Arcus Biosciences, Inc.: Current Employment. Ryba: Arcus Biosciences, Inc.: Current Employment. Scharville: Arcus Biosciences, Inc.: Current Employment. El-Baghdady: Arcus Biosciences, Inc.: Current Employment. OffLabel Disclosure: AB308 and zimberelimab (AB122) are in clinical development and are not approved in the United States for the use under discussion.


2005 ◽  
Vol 4 (4) ◽  
pp. 393-405 ◽  
Author(s):  
Wolfgang A. Bethge ◽  
Brenda M. Sandmaier

Radioimmunotherapy (RIT) combines the advantages of targeted radiation therapy and specific immunotherapy using monoclonal antibodies. RIT can be used either to target tumor cells or to specifically suppress immunocompetent host cells in the setting of allogeneic transplantation. The choice of radionuclide used for RIT depends on its distinct radiation characteristics and the type of malignancy or cells targeted. Beta-emitters with their lower energy and longer path length are more suitable to target bulky, solid tumors whereas α-emitters with their high linear energy transfer and short path length are better suited to target hematopoietic cells (normal or malignant). Different approaches of RIT such as the use of stable radioimmunoconjugates or of pretargeting strategies are available. Encouraging results have been obtained with RIT in patients with hematologic malignancies. The results in solid tumors are somewhat less favorable but new strategies for patients with minimal residual disease using adjuvant and locoregional treatment are evolving. This report outlines basic principles of RIT, gives an overview of available radionuclides and radioimmunoconjugates, and discusses clinical results with special emphasis on their use in hematologic malignancies including use in conditioning regimens for bone marrow transplantation.


2020 ◽  
Vol 04 (04) ◽  
pp. 345-350
Author(s):  
Ryan J. Slovak ◽  
Hyun S. Kim

AbstractThe reinfusion of autologous or allogeneic immune cells that have been educated and/or engineered ex vivo to respond to tumor-specific antigens is termed “adoptive cell therapy.” While adoptive cell therapy has made tremendous strides in the treatment of hematologic malignancies, its utilization for solid tumors has lagged somewhat behind. The purpose of this article is to concisely review the clinical research that has been done to investigate adoptive cell therapy as a treatment for gastrointestinal malignancies.


2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Julien Edeline ◽  
Roch Houot ◽  
Aurélien Marabelle ◽  
Marion Alcantara

AbstractChimeric antigen receptor (CAR)-modified T cells and BiTEs are both immunotherapies which redirect T cell specificity against a tumor-specific antigen through the use of antibody fragments. They demonstrated remarkable efficacy in B cell hematologic malignancies, thus paving the way for their development in solid tumors. Nonetheless, the use of such new drugs to treat solid tumors is not straightforward. So far, the results from early phase clinical trials are not as impressive as expected but many improvements are under way. In this review we present an overview of the clinical development of CAR-T cells and BiTEs targeting the main antigens expressed by solid tumors. We emphasize the most frequent hurdles encountered by either CAR-T cells or BiTEs, or both, and summarize the strategies that have been proposed to overcome these obstacles.


2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Sizhe Liu ◽  
Vasiliy Galat ◽  
Yekaterina Galat4 ◽  
Yoo Kyung Annie Lee ◽  
Derek Wainwright ◽  
...  

AbstractNatural killer (NK) cell is a specialized immune effector cell type that plays a critical role in immune activation against abnormal cells. Different from events required for T cell activation, NK cell activation is governed by the interaction of NK receptors with target cells, independent of antigen processing and presentation. Due to relatively unsophisticated cues for activation, NK cell has gained significant attention in the field of cancer immunotherapy. Many efforts are emerging for developing and engineering NK cell-based cancer immunotherapy. In this review, we provide our current understandings of NK cell biology, ongoing pre-clinical and clinical development of NK cell-based therapies and discuss the progress, challenges, and future perspectives.


Sign in / Sign up

Export Citation Format

Share Document