Anca Chelariu-Raicu, Sarina A Piha-Paul, Mariana Chavez-MacGregor, Jason Johnson, Raymond Sawaya, Mary Frances McAleer, Alissa Nguyen, Audrey Hartnett, Apostolia M Tsimberidou, Funda Meric-Bernstam, Ecaterina E Dumbrava
{"title":"Multidisciplinary Care of a Large Brain Metastasis in a Patient with Hormone-Receptor-Positive Breast Cancer with Ataxia-Telangiectasia Mutation.","authors":"Anca Chelariu-Raicu, Sarina A Piha-Paul, Mariana Chavez-MacGregor, Jason Johnson, Raymond Sawaya, Mary Frances McAleer, Alissa Nguyen, Audrey Hartnett, Apostolia M Tsimberidou, Funda Meric-Bernstam, Ecaterina E Dumbrava","doi":"10.36401/JIPO-22-33","DOIUrl":"https://doi.org/10.36401/JIPO-22-33","url":null,"abstract":"<p><p>Poly (adenosine diphosphate-ribose) polymerase inhibitors (PARP)i are emerging as standard oncology treatments in various tumor types. The indications will expand as PARPi are being investigated in various breast cancer subtypes. Currently, except <i>for BRCA1/2</i> mutation carriers with human epidermal growth factor receptor 2 (HER2)-negative breast cancer, there is inadequate identification of predictive biomarkers of response. We present a 57-year-old woman with metastatic breast cancer, hormone-receptor-positive, HER2 negative with a germline ataxia-telangiectasia mutation with a large brain metastasis with clinical benefit to talazoparib. This case report exemplifies the importance of the multidisciplinary management of patients with brain metastases and personalized biomarker selected treatment.</p>","PeriodicalId":16081,"journal":{"name":"Journal of Immunotherapy and Precision Oncology","volume":"6 3","pages":"158-161"},"PeriodicalIF":0.0,"publicationDate":"2023-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/bd/50/i2590-017X-6-3-158.PMC10448731.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10465136","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":"The Contemporary Landscape and Future Directions of Intratumoral Immunotherapy.","authors":"Sebastian Brito-Orama, Rahul A Sheth","doi":"10.36401/JIPO-22-8","DOIUrl":"https://doi.org/10.36401/JIPO-22-8","url":null,"abstract":"<p><p>Systemically administered immunotherapies have revolutionized the care of patients with cancer; however, for many cancer types, most patients do not exhibit objective responses. Intratumoral immunotherapy is a burgeoning strategy that is designed to boost the effectiveness of cancer immunotherapies across the spectrum of malignancies. By locally administering immune-activating therapies into the tumor itself, immunosuppressive barriers in the tumor microenvironment can be broken. Moreover, therapies too potent for systemic delivery can be safely administered to target location to maximize efficacy and minimize toxicity. In order for these therapies to be effective, though, they must be effectively delivered into the target tumor lesion. In this review, we summarize the current landscape of intratumoral immunotherapies and highlight key concepts that influence intratumoral delivery, and by extension, efficacy. We also provide an overview of the breadth and depth of approved minimally invasive delivery devices that can be considered to improve delivery of intratumoral therapies.</p>","PeriodicalId":16081,"journal":{"name":"Journal of Immunotherapy and Precision Oncology","volume":"6 2","pages":"84-90"},"PeriodicalIF":0.0,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/3f/ea/i2590-017X-6-2-84.PMC10195020.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10224911","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}
Mauro Di Pilato, Yun Gao, Yi Sun, Amina Fu, Carina Grass, Thomas Seeholzer, Regina Feederle, Irina Mazo, Samuel W Kazer, Kevin Litchfield, Ulrich H von Andrian, Thorsten R Mempel, Russell W Jenkins, Daniel Krappmann, Peter Keller
{"title":"Translational Studies Using the MALT1 Inhibitor (<i>S</i>)-Mepazine to Induce Treg Fragility and Potentiate Immune Checkpoint Therapy in Cancer.","authors":"Mauro Di Pilato, Yun Gao, Yi Sun, Amina Fu, Carina Grass, Thomas Seeholzer, Regina Feederle, Irina Mazo, Samuel W Kazer, Kevin Litchfield, Ulrich H von Andrian, Thorsten R Mempel, Russell W Jenkins, Daniel Krappmann, Peter Keller","doi":"10.36401/JIPO-22-18","DOIUrl":"https://doi.org/10.36401/JIPO-22-18","url":null,"abstract":"<p><strong>Introduction: </strong>Regulatory T cells (Tregs) play a critical role in the maintenance of immune homeostasis but also protect tumors from immune-mediated growth control or rejection and pose a significant barrier to effective immunotherapy. Inhibition of MALT1 paracaspase activity can selectively reprogram immune-suppressive Tregs in the tumor microenvironment to adopt a proinflammatory fragile state, which offers an opportunity to impede tumor growth and enhance the efficacy of immune checkpoint therapy (ICT).</p><p><strong>Methods: </strong>We performed preclinical studies with the orally available allosteric MALT1 inhibitor (<i>S</i>)-mepazine as a single-agent and in combination with anti-programmed cell death protein 1 (PD-1) ICT to investigate its pharmacokinetic properties and antitumor effects in several murine tumor models as well as patient-derived organotypic tumor spheroids (PDOTS).</p><p><strong>Results: </strong>(<i>S</i>)-mepazine demonstrated significant antitumor effects and was synergistic with anti-PD-1 therapy in vivo and ex vivo but did not affect circulating Treg frequencies in healthy rats at effective doses. Pharmacokinetic profiling revealed favorable drug accumulation in tumors to concentrations that effectively blocked MALT1 activity, potentially explaining preferential effects on tumor-infiltrating over systemic Tregs.</p><p><strong>Conclusions: </strong>The MALT1 inhibitor (<i>S</i>)-mepazine showed single-agent anticancer activity and presents a promising opportunity for combination with PD-1 pathway-targeted ICT. Activity in syngeneic tumor models and human PDOTS was likely mediated by induction of tumor-associated Treg fragility. This translational study supports ongoing clinical investigations (ClinicalTrials.gov Identifier: NCT04859777) of MPT-0118, (<i>S</i>)-mepazine succinate, in patients with advanced or metastatic treatment-refractory solid tumors.</p>","PeriodicalId":16081,"journal":{"name":"Journal of Immunotherapy and Precision Oncology","volume":"6 2","pages":"61-73"},"PeriodicalIF":0.0,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/06/11/i2590-017X-6-2-61.PMC10195017.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10208482","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}
Mirella Nardo, Deniz Can Guven, Aysun Senturk Yikilmaz, Shambhavi Singh, Jibran Ahmed
{"title":"Learning From Failure: Negative Trials in Oncology.","authors":"Mirella Nardo, Deniz Can Guven, Aysun Senturk Yikilmaz, Shambhavi Singh, Jibran Ahmed","doi":"10.36401/JIPO-23-X1","DOIUrl":"https://doi.org/10.36401/JIPO-23-X1","url":null,"abstract":"Learning From Failure: Negative Trials in Oncology Mirella Nardo, Deniz Can Guven, Aysun Senturk Yikilmaz, Shambhavi Singh, Jibran Ahmed Department of Investigational Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkey Department of Hematology, Denizli Government Hospital, Denizli, Turkey Department of Precision Oncology, Sir H N Reliance Foundation Hospital, Mumbai, India","PeriodicalId":16081,"journal":{"name":"Journal of Immunotherapy and Precision Oncology","volume":"6 2","pages":"59-60"},"PeriodicalIF":0.0,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/85/1f/i2590-017X-6-2-59.PMC10195015.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10224912","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":"Abstracts Presented at the 2022 Emirates Oncology Conference: November 25-27, 2022, Abu Dhabi, United Arab Emirates.","authors":"Khalid Balaraj","doi":"10.36401/JIPO-22-X6","DOIUrl":"https://doi.org/10.36401/JIPO-22-X6","url":null,"abstract":"s Presented at the 2022 Emirates Oncology Conference November 25-27, 2022, Abu Dhabi, United Arab Emirates J Immunother Precis Oncol. 2023; 6:117–126. DOI: 10.36401/JIPO-22-X6. This work is published under a CC-BY-NC-ND 4.0 International License. The Emirates Oncology Conference (EOC), now in its 10th year, continues to highlight important developments in the fields of cancer prevention and treatment. The Abu Dhabi Health Services SEHA and Tawam Hospital hosted the conference. These organizations are dedicated to offering the UAE top-tier healthcare and ongoing medical education to keep the public informed and ensure that medical experts remain up to date in their fields. The EOC featured significant sessions covering a wide range of topics with globally renowned speakers who kept the audience interested with their illuminating lectures and visual presentations. Breast cancer, hematological malignancies, palliative care, lung cancer, radiation oncology, pediatric oncology, genitourinary, gastrointestinal, and neuro-oncology were some of the topics covered. Over the course of the 3 days, there were about 4000 attendees including physicians, surgeons, researchers, healthcare professionals, and industry representatives from over 40 nations, of which 65% were from the UAE and 35% traveled from the USA, Europe, Middle East, and GCC. Approximately 150 speakers were hosted by EOC; speakers came from the USA, UK, Italy, Spain, France, Germany, Belgium, India, Pakistan, Philippines, Malaysia, Canada, and many regional nations such as Jordan, KSA, Oman, Bahrain, Lebanon, Egypt, and the UAE. In addition, more than 2 dozen abstracts were presented as oral and poster presentations, and the top 14 were chosen to be published in a medical journal as a result of EOC 2022. Selected abstracts are included herein. We are grateful to all members of the scientific and medical community, our organizing team, speakers, delegates and sponsors for their time, efforts, and contributions to making EOC one of the most important scientific events of the year. Dr. Kalid Balaraj Chair of Oncology Centre Tawam Hospital, UAE Journal of Immunotherapy and Precision Oncology 2023 | Volume 6 | Issue 2 | 117 jipoonline.org 118 EOC 2022 Abstracts Conference Proceedings 119 120 EOC 2022 Abstracts Conference Proceedings 121 122 EOC 2022 Abstracts Conference Proceedings 123 124 EOC 2022 Abstracts","PeriodicalId":16081,"journal":{"name":"Journal of Immunotherapy and Precision Oncology","volume":"6 2","pages":"117-126"},"PeriodicalIF":0.0,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/0b/ad/i2590-017X-6-2-117.PMC10195016.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9505549","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}
Zainab Fatima, Abdulrahman Abonofal, Bettzy Stephen
{"title":"Targeting Cancer Metabolism to Improve Outcomes with Immune Checkpoint Inhibitors.","authors":"Zainab Fatima, Abdulrahman Abonofal, Bettzy Stephen","doi":"10.36401/JIPO-22-27","DOIUrl":"https://doi.org/10.36401/JIPO-22-27","url":null,"abstract":"<p><p>Immune checkpoint inhibitors have revolutionized the treatment paradigm of several cancers. However, not all patients respond to treatment. Tumor cells reprogram metabolic pathways to facilitate growth and proliferation. This shift in metabolic pathways creates fierce competition with immune cells for nutrients in the tumor microenvironment and generates by-products harmful for immune cell differentiation and growth. In this review, we discuss these metabolic alterations and the current therapeutic strategies to mitigate these alterations to metabolic pathways that can be used in combination with checkpoint blockade to offer a new path forward in cancer management.</p>","PeriodicalId":16081,"journal":{"name":"Journal of Immunotherapy and Precision Oncology","volume":"6 2","pages":"91-102"},"PeriodicalIF":0.0,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/41/b7/i2590-017X-6-2-91.PMC10195018.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9848734","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}
Ian J Robertson, Timothy A Gregory, Steven G Waguespack, Marta Penas-Prado, Nazanin K Majd
{"title":"Recent Therapeutic Advances in Pituitary Carcinoma.","authors":"Ian J Robertson, Timothy A Gregory, Steven G Waguespack, Marta Penas-Prado, Nazanin K Majd","doi":"10.36401/JIPO-22-25","DOIUrl":"https://doi.org/10.36401/JIPO-22-25","url":null,"abstract":"<p><p>Pituitary carcinoma (PC) is a rare, aggressive malignancy that comprises 0.1-0.2% of all pituitary tumors. PC is defined anatomically as a pituitary tumor that metastasizes outside the primary intrasellar location as noncontiguous lesions in the central nervous system or as metastases to other organs. Similar to pituitary adenoma, PC originates from various cell types of the pituitary gland and can be functioning or nonfunctioning, with the former constituting the majority of the cases. Compression of intricate skull-based structures, excessive hormonal secretion, impaired pituitary function from therapy, and systemic metastases lead to debilitating symptoms and a poor survival outcome in most cases. PC frequently recurs despite multimodality treatments, including surgical resection, radiotherapy, and biochemical and cytotoxic treatments. There is an unmet need to better understand the pathogenesis and molecular characterization of PC to improve therapeutic strategies. As our understanding of the role of signaling pathways in the tumorigenesis of and malignant transformation of PC evolves, efforts have focused on targeted therapy. In addition, recent advances in the use of immune checkpoint inhibitors to treat various solid cancers have led to an interest in exploring the role of immunotherapy for the treatment of aggressive refractory pituitary tumors. Here, we review our current understanding of the pathogenesis, molecular characterization, and treatment of PC. Particular attention is given to emerging treatment options, including targeted therapy, immunotherapy, and peptide receptor radionuclide therapy.</p>","PeriodicalId":16081,"journal":{"name":"Journal of Immunotherapy and Precision Oncology","volume":"6 2","pages":"74-83"},"PeriodicalIF":0.0,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/14/f7/i2590-017X-6-2-74.PMC10195013.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10623392","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}
Felipe Soto, Luis F Torre-Sada, Frank E Mott, Sang T Kim, Roza Nurieva, Vickie R Shannon, Saadia A Faiz, Roberto F Casal, Mehmet Altan, Julie Lin, Ajay Sheshadri
{"title":"Sarcoidosis and Airway Disease After Immune Checkpoint Inhibitor Therapy: Case Study and Review of the Literature.","authors":"Felipe Soto, Luis F Torre-Sada, Frank E Mott, Sang T Kim, Roza Nurieva, Vickie R Shannon, Saadia A Faiz, Roberto F Casal, Mehmet Altan, Julie Lin, Ajay Sheshadri","doi":"10.36401/JIPO-22-30","DOIUrl":"10.36401/JIPO-22-30","url":null,"abstract":"<p><p>Pulmonary toxicity from immune checkpoint inhibitor therapy is typically a severe and potentially fatal complication, but these observations are driven by the most common toxicity, pneumonitis. Rarer pulmonary immune related adverse events, like airway disease and sarcoidosis, may have a more benign course. In this case report, we present a patient in whom therapy with the PD-1 inhibitor pembrolizumab resulted in severe eosinophilic asthma and sarcoidosis. This is the first case showing that anti-IL-5 inhibition may be safe in patients who develop eosinophilic asthma after ICI therapy. We further show that sarcoidosis does not necessarily require treatment cessation. This case highlights relevant nuances when clinicians face pulmonary toxicities other than pneumonitis.</p>","PeriodicalId":16081,"journal":{"name":"Journal of Immunotherapy and Precision Oncology","volume":"6 2","pages":"111-116"},"PeriodicalIF":0.0,"publicationDate":"2023-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/7e/d7/i2590-017X-6-2-111.PMC10195014.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10224914","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}
Juan I Ruiz, Maria A Lopez-Olivo, Yimin Geng, Maria E Suarez-Almazor
{"title":"COVID-19 Outcomes in Patients with Cancer Receiving Immune Checkpoint Inhibitors: A Systematic Review.","authors":"Juan I Ruiz, Maria A Lopez-Olivo, Yimin Geng, Maria E Suarez-Almazor","doi":"10.36401/JIPO-22-24","DOIUrl":"10.36401/JIPO-22-24","url":null,"abstract":"<p><strong>Introduction: </strong>Immune checkpoint inhibitors (ICIs) can cause inflammatory and immune-related adverse events (irAEs) that might worsen the course of COVID-19. We conducted a systematic review (PROSPERO ID: CRD42022307545) to evaluate the clinical course and complications of COVID-19 in patients with cancer receiving ICI.</p><p><strong>Methods: </strong>We searched Medline and Embase through January 5, 2022. We included studies evaluating patients with cancer who received ICI and developed COVID-19. Outcomes included mortality, severe COVID-19, intensive care unit (ICU) and hospital admissions, irAEs, and serious adverse events. We pooled data with random effects meta-analysis.</p><p><strong>Results: </strong>Twenty-five studies met study eligibility (<i>n</i> = 36,532 patients: 15,497 had COVID-19 and 3220 received ICI). Most studies (71.4%) had a high risk of comparability bias. There were no significant differences in mortality (relative risk [RR] 1.29; 95% CI 0.62-2.69), ICU admission (RR 1.20; 95% CI 0.71-2.00), and hospital admission (RR 0.91; 95% CI 0.79-1.06) when comparing patients treated with ICI with patients without cancer treatment. When pooling adjusted odds ratios (ORs), no statistically significant differences were observed in mortality (OR 0.95; 95% CI 0.57-1.60), severe COVID-19 (OR 1.05; 95% CI 0.45-2.46), or hospital admission (OR 2.02; 95% CI 0.96-4.27), when comparing patients treated with ICIs versus patients with cancer without ICI therapy. No significant differences were observed when comparing clinical outcomes in patients receiving ICIs versus patients receiving any of the other anticancer therapies.</p><p><strong>Conclusion: </strong>Although current evidence is limited, COVID-19 clinical outcomes of patients with cancer receiving ICI therapy appear to be similar to those not receiving oncologic treatment or other cancer therapies.</p>","PeriodicalId":16081,"journal":{"name":"Journal of Immunotherapy and Precision Oncology","volume":"6 2","pages":"103-110"},"PeriodicalIF":0.0,"publicationDate":"2023-02-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/6b/6e/i2590-017X-6-2-103.PMC10195019.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9848735","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":"Therapeutic Targeting of DNA Damage Repair in the Era of Precision Oncology and Immune Checkpoint Inhibitors.","authors":"Curtis A Clark, Eddy S Yang","doi":"10.36401/JIPO-22-15","DOIUrl":"https://doi.org/10.36401/JIPO-22-15","url":null,"abstract":"<p><p>Cancer manifestation is a multistep process involving accumulation of various genetic and epigenetic changes that results in oncogenic \"hallmarks of cancer\" processes including genomic instability. Exploitation of aberrant DNA-damage response (DDR) mechanisms in cancer is in part a goal of many therapeutic strategies, and recent evidence supports the role of targeting DDR in modulating the tumor immune microenvironment to enhance immunotherapeutic response. Improved cancer profiling, including next-generation and whole-genome mutational sequencing of tumor tissue, as well as circulating nucleic acids, has enhanced our understanding of the genetic and epigenetic molecular mechanisms in tumorigenesis and will become fundamental to precisely target tumors and achieve cancer control. With the successes of poly(ADP-ribose) polymerase inhibitors (PARPi) and immunotherapies, the intersection of DDR molecular machinery and corresponding antitumor immune response has gained much interest with a focus on achieving therapeutic synergy using DNA damage-targeting agents and immunotherapy. In this review, we provide a bench-to-bedside overview of the fundamentals of DDR signaling and repair as they relate to cancer therapeutic strategies including novel DDR-targeting agents. We also discuss the underlying mechanisms that link DDR signaling to antitumor immunity and immunotherapy efficacy, and how this knowledge can be used to improve precision medicine approaches in the treatment of cancer.</p>","PeriodicalId":16081,"journal":{"name":"Journal of Immunotherapy and Precision Oncology","volume":"6 1","pages":"31-49"},"PeriodicalIF":0.0,"publicationDate":"2023-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/74/a1/i2590-017X-6-1-31.PMC9888518.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10674938","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}