Immunotherapy advancesPub Date : 2022-04-08eCollection Date: 2022-01-01DOI: 10.1093/immadv/ltac008
Guilherme Ferreira de Britto Evangelista, Amanda Braga Figueiredo, Milton José de Barros E Silva, Kenneth J Gollob
{"title":"Balancing the good and the bad: controlling immune-related adverse events versus anti-tumor responses in cancer patients treated with immune checkpoint inhibitors.","authors":"Guilherme Ferreira de Britto Evangelista, Amanda Braga Figueiredo, Milton José de Barros E Silva, Kenneth J Gollob","doi":"10.1093/immadv/ltac008","DOIUrl":"https://doi.org/10.1093/immadv/ltac008","url":null,"abstract":"<p><p>Immune checkpoint inhibitors (ICI) have provided new hope for cancer patients, and in particular for patients with tumors that are immunologically active and classified as hot tumors. These tumors express antigenic and tumor microenvironment (TME) characteristics that make them potential candidates for therapy with checkpoint inhibitors that aim to reactivate the immune response such as anti-PD-1 and anti-CTLA-4. Examples of potentially responsive cancers are, melanoma, non-small cell lung cancer and several other metastatic or unresectable tumors with genetic instability: DNA mismatch repair deficiency (dMMR), microsatellite instability-high (MSI-H), or with a high tumor mutational burden (TMB). Immunotherapy using checkpoint inhibitors is typically associated with adverse events (AEs) that are milder than those with chemotherapy. However, a significant percentage of patients develop short-term immune-related AEs (irAEs) which range from mild (~70%) to severe cases (~13%) that can lead to modifications of the checkpoint inhibitor therapy and in some cases, death. While some studies have investigated immune mechanisms behind the development of irAEs, much more research is needed to understand the mechanisms and to develop interventions that could attenuate severe irAEs, while maintaining the anti-tumor response intact. Moreover, studies to identify biomarkers that can predict the likelihood of a patient developing severe irAEs would be of great clinical importance. Here we discuss some of the clinical ramifications of irAEs, potential immune mechanisms behind their development and studies that have investigated potentially useful biomarkers of irAEs development.</p>","PeriodicalId":73353,"journal":{"name":"Immunotherapy advances","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-04-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9327097/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40687874","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Immunotherapy advancesPub Date : 2022-02-21eCollection Date: 2022-01-01DOI: 10.1093/immadv/ltac007
A Ammon, L Mellenthin, C Emmerich, E Naschberger, M Stürzl, A Mackensen, F Müller
{"title":"Reduced cytotoxicity by mutation of lysine 590 of <i>Pseudomonas</i> exotoxin can be restored in an optimized, lysine-free immunotoxin.","authors":"A Ammon, L Mellenthin, C Emmerich, E Naschberger, M Stürzl, A Mackensen, F Müller","doi":"10.1093/immadv/ltac007","DOIUrl":"https://doi.org/10.1093/immadv/ltac007","url":null,"abstract":"<p><p>Immunotoxins, which are fusion proteins of an antibody fragment and a fragment of a bacterial or a plant toxin, induce apoptosis in target cells by inhibition of protein synthesis. ADP-ribosylating toxins often have few lysine residues in their catalytic domain. As they are the target for ubiquitination, the low number of lysines possibly prevents ubiquitin-dependent degradation of the toxin in the cytosol. To reduce this potential degradation, we aimed to generate a lysine-free (noK), <i>Pseudomonas</i> exotoxin (PE)-based immunotoxin. The new generation 24 kDa PE, which lacks all but the furin-cleavage site of domain II, was mutated at lysine 590 (K590) and at K606 in a CD22-targeting immunotoxin and activity was determined against various B cell malignancies <i>in vitro</i> and <i>in vivo</i>. On average, K590 mutated to arginine (R) reduced cytotoxicity by 1.3-fold and K606R enhanced cytotoxicity by 1.3-fold compared to <i>wild type</i> (<i>wt</i>). Mutating K590 to histidine or deleting K590 did not prevent this loss in cytotoxicity. Neither stability nor internalization rate of K590R could explain reduced cytotoxicity. These results highlight the relevance of lysine 590 for PE intoxication. In line with <i>in vitro</i> results, the K606R mutant was more than 1.8-fold more active than the other variants <i>in vivo</i> suggesting that this single mutation may be beneficial when targeting CD22-positive malignancies. Finally, reduced cytotoxicity by K590R was compensated for by K606R and the resulting lysine-free variant achieved <i>wt</i>-like activity <i>in vitro</i> and <i>in vivo</i>. Thus, PE24-noK may represent a promising candidate for down-stream applications that would interfere with lysines.</p>","PeriodicalId":73353,"journal":{"name":"Immunotherapy advances","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-02-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9327129/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40666626","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"New and emerging concepts and therapies for the treatment of food allergy","authors":"David W Hwang, C. Nagler, C. Ciaccio","doi":"10.1093/immadv/ltac006","DOIUrl":"https://doi.org/10.1093/immadv/ltac006","url":null,"abstract":"Abstract Food allergy is an increasingly common disease that often starts in early childhood and lasts throughout life. Self-reported food allergy has risen at a rate of 1.2% per decade since 1988, and by 2018, the prevalence of food allergy in the United States was estimated to be 8% in children and 11% in adults.- This prevalence has led to an economic burden of almost $25 billion annually. Despite these staggering statistics, as of the time of this writing, the Food and Drug Administration (FDA) has only approved one treatment for food allergy, which is limited to use in children with peanut allergy. Fortunately, a new horizon of therapeutic interventions, in all stages of development, lay ahead and hold promise for the near future.","PeriodicalId":73353,"journal":{"name":"Immunotherapy advances","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-02-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"46430771","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Immunotherapy advancesPub Date : 2022-01-31eCollection Date: 2022-01-01DOI: 10.1093/immadv/ltac004
Simone Reinwald, Jennifer M Rolland, Robyn E O'Hehir, Menno C van Zelm
{"title":"Peanut oral immunotherapy: current trends in clinical trials.","authors":"Simone Reinwald, Jennifer M Rolland, Robyn E O'Hehir, Menno C van Zelm","doi":"10.1093/immadv/ltac004","DOIUrl":"https://doi.org/10.1093/immadv/ltac004","url":null,"abstract":"<p><p>Immunotherapy for allergy has been practiced for over 100 years. Low-dose repeated exposure to specific allergen extracts over several months to years can successfully induce clinical tolerance in patients with allergy to insect venoms, pollen, house dust mite, and domestic animals. Different regimens and routes for immunotherapy include subcutaneous, sublingual, oral, and intralymphatic. Food allergies have been difficult to treat in this way due to high anaphylactic potential and only recently the first immunotherapy for peanut allergy has received regulatory approval. Several clinical trials have indicated high efficacy in desensitisation of peanut-allergic individuals using oral immunotherapy, which allows for safer administration of relatively high allergen concentrations. Still, the risk of adverse events including serious allergic reactions and high anxiety levels for patients remains, demonstrating the need for further optimisation of treatment protocols. Here we discuss the design and outcomes of recent clinical trials with traditional oral immunotherapy, and consider alternative protocols and formulations for safer and more effective oral treatment strategies for peanut allergy.</p>","PeriodicalId":73353,"journal":{"name":"Immunotherapy advances","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-01-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9327116/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40666627","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Immunotherapy advancesPub Date : 2022-01-27eCollection Date: 2022-01-01DOI: 10.1093/immadv/ltac002
D Tatovic, M A McAteer, J Barry, A Barrientos, K Rodríguez Terradillos, I Perera, E Kochba, Y Levin, M Dul, S A Coulman, J C Birchall, C von Ruhland, A Howell, R Stenson, M Alhadj Ali, S D Luzio, G Dunseath, W Y Cheung, G Holland, K May, J R Ingram, M M U Chowdhury, F S Wong, R Casas, C Dayan, J Ludvigsson
{"title":"Safety of the use of gold nanoparticles conjugated with proinsulin peptide and administered by hollow microneedles as an immunotherapy in type 1 diabetes.","authors":"D Tatovic, M A McAteer, J Barry, A Barrientos, K Rodríguez Terradillos, I Perera, E Kochba, Y Levin, M Dul, S A Coulman, J C Birchall, C von Ruhland, A Howell, R Stenson, M Alhadj Ali, S D Luzio, G Dunseath, W Y Cheung, G Holland, K May, J R Ingram, M M U Chowdhury, F S Wong, R Casas, C Dayan, J Ludvigsson","doi":"10.1093/immadv/ltac002","DOIUrl":"https://doi.org/10.1093/immadv/ltac002","url":null,"abstract":"<p><p>Antigen-specific immunotherapy is an immunomodulatory strategy for autoimmune diseases, such as type 1 diabetes, in which patients are treated with autoantigens to promote immune tolerance, stop autoimmune β-cell destruction and prevent permanent dependence on exogenous insulin. In this study, human proinsulin peptide C19-A3 (known for its positive safety profile) was conjugated to ultrasmall gold nanoparticles (GNPs), an attractive drug delivery platform due to the potential anti-inflammatory properties of gold. We hypothesised that microneedle intradermal delivery of C19-A3 GNP may improve peptide pharmacokinetics and induce tolerogenic immunomodulation and proceeded to evaluate its safety and feasibility in a first-in-human trial. Allowing for the limitation of the small number of participants, intradermal administration of C19-A3 GNP appears safe and well tolerated in participants with type 1 diabetes. The associated prolonged skin retention of C19-A3 GNP after intradermal administration offers a number of possibilities to enhance its tolerogenic potential, which should be explored in future studies.</p>","PeriodicalId":73353,"journal":{"name":"Immunotherapy advances","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-01-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/8b/e2/ltac002.PMC9327128.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40666629","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Immunotherapy advancesPub Date : 2022-01-25eCollection Date: 2022-01-01DOI: 10.1093/immadv/ltac005
Rafaela Rossetti, Heloísa Brand, Sarah Caroline Gomes Lima, Izadora Peter Furtado, Roberta Maraninchi Silveira, Daianne Maciely Carvalho Fantacini, Dimas Tadeu Covas, Lucas Eduardo Botelho de Souza
{"title":"Combination of genetically engineered T cells and immune checkpoint blockade for the treatment of cancer.","authors":"Rafaela Rossetti, Heloísa Brand, Sarah Caroline Gomes Lima, Izadora Peter Furtado, Roberta Maraninchi Silveira, Daianne Maciely Carvalho Fantacini, Dimas Tadeu Covas, Lucas Eduardo Botelho de Souza","doi":"10.1093/immadv/ltac005","DOIUrl":"https://doi.org/10.1093/immadv/ltac005","url":null,"abstract":"<p><p>Immune checkpoint (IC) blockade using monoclonal antibodies is currently one of the most successful immunotherapeutic interventions to treat cancer. By reinvigorating antitumor exhausted T cells, this approach can lead to durable clinical responses. However, the majority of patients either do not respond or present a short-lived response to IC blockade, in part due to a scarcity of tumor-specific T cells within the tumor microenvironment. Adoptive transfer of T cells genetically engineered to express chimeric antigen receptors (CARs) or engineered T-cell receptors (TCRs) provide the necessary tumor-specific immune cell population to target cancer cells. However, this therapy has been considerably ineffective against solid tumors in part due to IC-mediated immunosuppressive effects within the tumor microenvironment. These limitations could be overcome by associating adoptive cell transfer of genetically engineered T cells and IC blockade. In this comprehensive review, we highlight the strategies and outcomes of preclinical and clinical attempts to disrupt IC signaling in adoptive T-cell transfer against cancer. These strategies include combined administration of genetically engineered T cells and IC inhibitors, engineered T cells with intrinsic modifications to disrupt IC signaling, and the design of CARs against IC molecules. The current landscape indicates that the synergy of the fast-paced refinements of gene-editing technologies and synthetic biology and the increased comprehension of IC signaling will certainly translate into a novel and more effective immunotherapeutic approaches to treat patients with cancer.</p>","PeriodicalId":73353,"journal":{"name":"Immunotherapy advances","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-01-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/1e/14/ltac005.PMC9327125.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40666623","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Immunotherapy advancesPub Date : 2022-01-11eCollection Date: 2022-01-01DOI: 10.1093/immadv/ltac001
Tim Elliott
{"title":"Immunotherapy advances: One year on.","authors":"Tim Elliott","doi":"10.1093/immadv/ltac001","DOIUrl":"10.1093/immadv/ltac001","url":null,"abstract":"","PeriodicalId":73353,"journal":{"name":"Immunotherapy advances","volume":null,"pages":null},"PeriodicalIF":4.1,"publicationDate":"2022-01-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/1f/c5/ltac001.PMC9327108.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40666625","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Stephanie Schwartz, Nidhi Patel, Tyler Longmire, Pushpa Jayaraman, Xiaomo Jiang, Hongbo Lu, Lisa Baker, Janelle Velez, Radha Ramesh, Anne-Sophie Wavreille, Melanie Verneret, Hong Fan, Tiancen Hu, Fangmin Xu, John Taraszka, Marc Pelletier, Joy Miyashiro, Mikael Rinne, Glenn Dranoff, Catherine Sabatos-Peyton, Viviana Cremasco
{"title":"Characterization of sabatolimab, a novel immunotherapy with immuno-myeloid activity directed against TIM-3 receptor.","authors":"Stephanie Schwartz, Nidhi Patel, Tyler Longmire, Pushpa Jayaraman, Xiaomo Jiang, Hongbo Lu, Lisa Baker, Janelle Velez, Radha Ramesh, Anne-Sophie Wavreille, Melanie Verneret, Hong Fan, Tiancen Hu, Fangmin Xu, John Taraszka, Marc Pelletier, Joy Miyashiro, Mikael Rinne, Glenn Dranoff, Catherine Sabatos-Peyton, Viviana Cremasco","doi":"10.1093/immadv/ltac019","DOIUrl":"https://doi.org/10.1093/immadv/ltac019","url":null,"abstract":"<p><strong>Objectives: </strong>Sabatolimab is a humanized monoclonal antibody (hIgG4, S228P) directed against human T-cell immunoglobulin domain and mucin domain-3 (TIM-3). Herein, we describe the development and characterization of sabatolimab.</p><p><strong>Methods: </strong>Sabatolimab was tested for binding to its target TIM-3 and blocking properties. The functional effects of sabatolimab were tested in T-cell killing and myeloid cell cytokine assays. Antibody-mediated cell phagocytosis (ADCP) by sabatolimab was also assessed.</p><p><strong>Results: </strong>Sabatolimab was shown to (i) enhance T-cell killing and inflammatory cytokine production by dendritic cells (DCs); (ii) facilitate the phagocytic uptake of TIM-3-expressing target cells; and (iii) block the interaction between TIM-3 and its ligands PtdSer/galectin-9.</p><p><strong>Conclusion: </strong>Taken together, our results support both direct anti-leukemic effects and immune-mediated modulation by sabatolimab, reinforcing the notion that sabatolimab represents a novel immunotherapy with immuno-myeloid activity, holding promise for the treatment of myeloid cell neoplasms.</p>","PeriodicalId":73353,"journal":{"name":"Immunotherapy advances","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9525012/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10743395","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ashish K Marwaha, Samuel Chow, Anne M Pesenacker, Laura Cook, Annika Sun, S Alice Long, Jennie H M Yang, Kirsten A Ward-Hartstonge, Evangelia Williams, Clara Domingo-Vila, Khalif Halani, Kristina M Harris, Timothy I M Tree, Megan K Levings, Thomas Elliott, Rusung Tan, Jan P Dutz
{"title":"A phase 1b open-label dose-finding study of ustekinumab in young adults with type 1 diabetes.","authors":"Ashish K Marwaha, Samuel Chow, Anne M Pesenacker, Laura Cook, Annika Sun, S Alice Long, Jennie H M Yang, Kirsten A Ward-Hartstonge, Evangelia Williams, Clara Domingo-Vila, Khalif Halani, Kristina M Harris, Timothy I M Tree, Megan K Levings, Thomas Elliott, Rusung Tan, Jan P Dutz","doi":"10.1093/immadv/ltab022","DOIUrl":"https://doi.org/10.1093/immadv/ltab022","url":null,"abstract":"<p><strong>Objectives: </strong>We assessed the safety of ustekinumab (a monoclonal antibody used in psoriasis to target the IL-12 and IL-23 pathways) in a small cohort of recent-onset (<100 days of diagnosis) adults with type 1 diabetes (T1D) by conducting a pilot open-label dose-finding and mechanistic study (NCT02117765) at the University of British Columbia.</p><p><strong>Methods: </strong>We sequentially enrolled 20 participants into four subcutaneous dosing cohorts: (i) 45 mg loading weeks 0/4/16, (ii) 45 mg maintenance weeks 0/4/16/28/40, (iii) 90 mg loading weeks 0/4/16, and (iv) 90 mg maintenance weeks 0/4/16/28/40. The primary endpoint was safety as assessed by an independent data and safety monitoring board (DSMB) but we also measured mixed meal tolerance test C-peptide, insulin use/kg, and HbA1c. Immunophenotyping was performed to assess immune cell subsets and islet antigen-specific T cell responses.</p><p><strong>Results: </strong>Although several adverse events were reported, only two (bacterial vaginosis and hallucinations) were thought to be possibly related to drug administration by the study investigators. At 1 year, the 90 mg maintenance dosing cohort had the smallest mean decline in C-peptide area under the curve (AUC) (0.1 pmol/ml). Immunophenotyping showed that ustekinumab reduced the percentage of circulating Th17, Th1, and Th17.1 cells and proinsulin-specific T cells that secreted IFN-γ and IL-17A.</p><p><strong>Conclusion: </strong>Ustekinumab was deemed safe to progress to efficacy studies by the DSMB at doses used to treat psoriasis in adults with T1D. A 90 mg maintenance dosing schedule reduced proinsulin-specific IFN-γ and IL-17A-producing T cells. Further studies are warranted to determine if ustekinumab can prevent C-peptide AUC decline and induce a clinical response.</p>","PeriodicalId":73353,"journal":{"name":"Immunotherapy advances","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8769169/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9764729","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Dayane Schmidt, Sima Ebrahimabadi, Kauan Ribeiro de Sena Gomes, Graziela de Moura Aguiar, Mariane Cariati Tirapelle, Renata Nacasaki Silvestre, Júlia Teixeira Cottas de Azevedo, Dimas Tadeu Covas, Virginia Picanço-Castro
{"title":"Engineering CAR-NK cells: how to tune innate killer cells for cancer immunotherapy.","authors":"Dayane Schmidt, Sima Ebrahimabadi, Kauan Ribeiro de Sena Gomes, Graziela de Moura Aguiar, Mariane Cariati Tirapelle, Renata Nacasaki Silvestre, Júlia Teixeira Cottas de Azevedo, Dimas Tadeu Covas, Virginia Picanço-Castro","doi":"10.1093/immadv/ltac003","DOIUrl":"https://doi.org/10.1093/immadv/ltac003","url":null,"abstract":"<p><p>Cell therapy is an innovative approach that permits numerous possibilities in the field of cancer treatment. CAR-T cells have been successfully used in patients with hematologic relapsed/refractory. However, the need for autologous sources for T cells is still a major drawback. CAR-NK cells have emerged as a promising resource using allogeneic cells that could be established as an off-the-shelf treatment. NK cells can be obtained from various sources, such as peripheral blood (PB), bone marrow, umbilical cord blood (CB), and induced pluripotent stem cells (iPSC), as well as cell lines. Genetic engineering of NK cells to express different CAR constructs for hematological cancers and solid tumors has shown promising preclinical results and they are currently being explored in multiple clinical trials. Several strategies have been employed to improve CAR-NK-cell expansion and cytotoxicity efficiency. In this article, we review the latest achievements and progress made in the field of CAR-NK-cell therapy.</p>","PeriodicalId":73353,"journal":{"name":"Immunotherapy advances","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2022-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9327111/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10309720","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}