1
|
Sweatman E, Bayley R, Selemane R, Higgs MR. SETD1A-dependent EME1 transcription drives PARPi sensitivity in HR deficient tumour cells. Br J Cancer 2025; 132:690-702. [PMID: 39994444 PMCID: PMC11997087 DOI: 10.1038/s41416-025-02963-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 01/14/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Cells deficient in DNA repair factors breast cancer susceptibility 1/2 (BRCA1/2) or ataxia-telangiectasia mutated (ATM) are sensitive to poly-ADP ribose polymerase (PARP) inhibitors. Building on our previous findings, we asked how the lysine methyltransferase SETD1A contributed to PARP inhibitor-mediated cell death in these contexts and determined the mechanisms responsible. METHODS We used cervical, breast, lung and ovarian cancer cells bearing mutations in BRCA1 or ATM and depleted SETD1A using siRNA or CRISPR/Cas9. We assessed the effects of the PARPi Olaparib on cell viability, homologous recombination, and DNA repair. We assessed underlying transcriptional perturbations using RNAseq. We used The Cancer Genomics Atlas (TCGA) and DepMap to investigate patient survival and cancer cell characteristics. RESULTS Loss of SETD1A from both BRCA1-deficient and ATM-deficient cancer cells was associated with resistance to Olaparib, explained by partial restoration of homologous recombination. Mechanistically, SETD1A-dependent transcription of the crossover junction endonuclease EME1 correlated with sensitivity to Olaparib in these cells. Accordingly, when SETD1A or EME1 was lost, BRCA1 or ATM-mutated cells became resistant to Olaparib, and homologous recombination was partially restored. CONCLUSIONS Loss of SETD1A or EME1 drives cellular resistance to Olaparib in certain genetic contexts and may help explain why patients develop resistance to PARP inhibitors in the clinic.
Collapse
Affiliation(s)
- Ellie Sweatman
- Department of Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Rachel Bayley
- Department of Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Richad Selemane
- Department of Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Martin R Higgs
- Department of Cancer and Genomic Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK.
| |
Collapse
|
2
|
Schubart C, Tögel L, Carta MG, Hetzner P, Helbig L, Zaglas C, Ziegler M, Stöhr R, Hölsken A, Hoyer J, Ferrazzi F, Neufert C, Lettmaier S, Pavel M, Golcher H, Mueller SK, Fuchs F, Schulmeyer CE, Beckmann MW, Wullich B, Agaimy A, Reis A, Hartmann A, Meidenbauer N, Spoerl S, Haller F, Moskalev EA. Limited association between HRR gene alterations and HRD in molecular tumor board cancer samples: Who should be tested for HRD? Int J Cancer 2025. [PMID: 40278800 DOI: 10.1002/ijc.35457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 12/02/2024] [Accepted: 01/25/2025] [Indexed: 04/26/2025]
Abstract
Alterations in Homologous Recombination Repair (HRR) Pathway genes have been found to be associated with HR-Deficiency (HRD), which is an approved biomarker for PARP Inhibitor (PARPi) treatment. The aim of a Molecular Tumor Board (MTB) is to identify molecular alterations in cancer patients with advanced tumors that may suggest off-label treatment options. So far, few studies have analyzed the presence of HRR gene mutations and their association with HRD outside of clinical studies. Currently, no data on HRD testing in the setting of a MTB have been published. For the present study, a cohort of 237 patients encompassing 24 different tumor entities was collected from the MTB of the Comprehensive Cancer Center Erlangen-EMN. We show that an elevated Genomic Instability Score (GIS ≥42) can occur in samples with and without mutations in HRR-related genes. Overall, 38.1% of cancer samples with BRCA1/2 mutations, 10.9% of tumors with alterations in HRR genes other than BRCA1/2, and 4.3% of cancer samples without HRR gene mutations harbored an elevated GIS. Notably, our data show that various inactivating BRCA1/2 mutations are not associated with an elevated GIS. Taken together, panCancer assessment of HRD in addition to BRCA1/2 and other HRR gene mutational analysis is recommended to guide decisions regarding PARPi treatment. Further studies are needed to establish thresholds for GIS in non-ovarian cancer entities. Finally, HRD can be observed in 4.3% of BRCA1/2 and other HRR gene wildtype cancer samples, and may emerge as an independent biomarker for PARPi in the future.
Collapse
Affiliation(s)
- Christoph Schubart
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
| | - Lars Tögel
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
| | - Maria Giulia Carta
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
| | - Philip Hetzner
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lina Helbig
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Charlotte Zaglas
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Maria Ziegler
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Robert Stöhr
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
| | - Annett Hölsken
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
| | - Juliane Hoyer
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Fulvia Ferrazzi
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Department of Nephropathology, Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Clemens Neufert
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sebastian Lettmaier
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Marianne Pavel
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Henriette Golcher
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sarina K Mueller
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Department of ENT, Head and Neck Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Florian Fuchs
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Carla E Schulmeyer
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Department of Obstetrics and Gynecology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Matthias W Beckmann
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Department of Obstetrics and Gynecology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Bernd Wullich
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Department of Urology and Pediatric Urology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Abbas Agaimy
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
| | - Andre Reis
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Institute of Human Genetics, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Centre for Rare Diseases Erlangen, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
| | - Norbert Meidenbauer
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Department of Internal Medicine 5, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Silvia Spoerl
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
- Department of Internal Medicine 5, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Florian Haller
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA), Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany
- Zentrum Personalisierte Medizin Erlangen (ZPM-Erlangen), Erlangen, Germany
| | - Evgeny A Moskalev
- Institute of Pathology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
3
|
García-Díaz HC, Larrosa-Garcia M, Gómez-Alonso J, Cruellas M, Felip E, Macarulla T, Farriols A, Carreras MJ. Off-label use of olaparib in uncommon tumor locations in patients with impaired homologous recombination genes. FARMACIA HOSPITALARIA 2025:S1130-6343(25)00017-0. [PMID: 40155245 DOI: 10.1016/j.farma.2025.02.010] [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: 12/18/2024] [Revised: 01/28/2025] [Accepted: 02/18/2025] [Indexed: 04/01/2025] Open
Abstract
OBJECTIVE To describe the effectiveness and safety of olaparib off-label indications in patients with impaired homologous recombination genes and solid tumors different than those authorized. METHODS A single-center, observational and retrospective study including patients treated with olaparib for off-label use. The main variables were patient characteristics, prior therapies, response to therapy, progression-free survival, overall survival and adverse events. RESULTS A total of 6 patients were included. All patients had metastases and received 3 or more lines of prior treatment. The primary tumor locations and mutations were partner and localizer of BRCA2 (PALB2) intrahepatic cholangiocarcinoma, ataxia telangiectasia mutated (ATM) non-small cell lung adenocarcinoma, somatic breast cancer gene (sBRCA2) colorectal cancer, germinal breast cancer gene 2 (gBRCA2) breast neuroendocrine tumor, gBRCA2 ampullary cancer and gBRCA2 pancreatic neuroendocrine tumor. At the end of the study, one patient was still receiving olaparib showing more than 25 months of sustained stable disease response. No novel toxicities were observed besides those included in the product information. CONCLUSIONS There is limited published evidence on the use of olaparib in patients harboring pathogenic variants other than breast cancer genes, like PALB2 and ATM and conditions different than those authorized such as digestive tract, neuroendocrine and lung tumors. Further research is to assess the efficacy of olaparib in these patients.
Collapse
Affiliation(s)
| | | | - Javier Gómez-Alonso
- Department of Pharmacy, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Mara Cruellas
- Medical Oncology Department, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Enriqueta Felip
- Medical Oncology Department, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Teresa Macarulla
- Medical Oncology Department, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Anna Farriols
- Department of Pharmacy, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| | - Maria J Carreras
- Department of Pharmacy, Vall d'Hebron Hospital Universitari, Barcelona, Spain
| |
Collapse
|
4
|
Muzzana M, Broggini M, Damia G. The Landscape of PARP Inhibitors in Solid Cancers. Onco Targets Ther 2025; 18:297-317. [PMID: 40051775 PMCID: PMC11884256 DOI: 10.2147/ott.s499226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
PARP inhibitors are a class of agents that have shown significant preclinical activity in models defective in homologous recombination (HR). The identification of synthetic lethality between HR defects and PARP inhibition led to several clinical trials in tumors with known HR defects (initially mutations in BRCA1/2 genes and subsequently in other genes involved in HR). These studies demonstrated significant responses in breast and ovarian cancers, which are known to have a significant proportion of patients with HR defects. Since the approval of the first PARP inhibitor (PARPi), olaparib, several other inhibitors have been developed, expanding the armamentarium available to clinicians in this setting. The positive results obtained in breast and ovarian cancer have expanded the use of PARPi in other solid tumors with HR defects, including prostate and pancreatic cancer in which these defects have been identified. The clinical trials have demonstrated responses to PARPi which are now also available for the subset of patients with prostate and pancreatic cancer with HR defects. This review summarizes the results obtained in solid tumors with PARPi and their potential use when combined with other agents, including immune checkpoint inhibitors that are likely to further increase the survival of these patients which still needs a dramatic improvement.
Collapse
Affiliation(s)
- Marta Muzzana
- Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Massimo Broggini
- Experimental Oncology Department, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Giovanna Damia
- Experimental Oncology Department, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
5
|
Woodfield D, Le T, Prince G, Lee H, Vachhani H, Majety P. Use of Olaparib in the Management of Metastatic Parathyroid Carcinoma With BRCA1 Mutation. JCEM CASE REPORTS 2025; 3:luaf007. [PMID: 39963301 PMCID: PMC11831516 DOI: 10.1210/jcemcr/luaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Indexed: 02/20/2025]
Abstract
Parathyroid carcinoma (PC) is a rare cause of primary hyperparathyroidism with a highly variable clinical course. We report the case of a man with metastatic PC who presented with severe hypercalcemia, discovered incidentally after a fall. He underwent left upper parathyroidectomy with left thyroid lobectomy, and pathology confirmed PC. After a year of absence from follow-up, he developed recurrence with bilateral pulmonary metastases. Cinacalcet and denosumab were initiated due to persistent, severe hypercalcemia, followed by wedge resection and palliative radiotherapy of pulmonary metastases. Genetic analysis revealed no actionable pathogenic variants, but a BRCA1 mutation classified as a variant of unknown significance (VUS) was identified. He was started on olaparib, a poly adenosine diphosphate-ribose polymerase (PARP) inhibitor, 3 years after initial diagnosis. Following this, his PTH level declined by approximately 40% within 7 months. Subsequently, his PTH levels began increasing despite continuation of olaparib and, after 20 months, rose to his original PTH level prior to the initiation of therapy. This is a unique case of a patient with metastatic PC who had a BRCA1 VUS mutation, with initial partial reduction in PTH and calcium levels after PARP inhibitor treatment.
Collapse
Affiliation(s)
- David Woodfield
- Department of Endocrinology, Diabetes, and Metabolism, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Trang Le
- Department of Endocrinology, Diabetes, and Metabolism, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Grace Prince
- Department of Endocrinology, Diabetes, and Metabolism, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Hyun Lee
- Department of Hematology, Oncology, and Palliative Care, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Hetal Vachhani
- Department of Hematology, Oncology, and Palliative Care, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Priyanka Majety
- Department of Endocrinology, Diabetes, and Metabolism, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
6
|
Park W, Fricke J, Mambetsariev I, Velasquez G, Nadaf-Rahrov R, Dingal ST, Kim P, Babikian R, Amini A, Afkhami M, Salgia R. Novel LZTR1 germline mutation as a mechanism of resistance to osimertinib in EGFR-mutated lung adenocarcinoma: a case report. Transl Lung Cancer Res 2025; 14:625-630. [PMID: 40114953 PMCID: PMC11921366 DOI: 10.21037/tlcr-24-723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/30/2024] [Indexed: 03/22/2025]
Abstract
Background Tyrosine kinase inhibitors (TKIs) are now the standard of care first-line therapy for epidermal growth factor receptor (EGFR)-mutated advanced non-small cell lung cancer (NSCLC) patients. Despite positive outcomes in most patients, with extended progression-free survival (PFS), a small population of patients respond poorly to these drugs. Complex genetic and non-genetic resistance mechanisms may be the drivers of disease in cancer, but further research is required to identify these mechanisms in the clinic. Germline molecular testing alongside broad-panel somatic next-generation sequencing (NGS) has allowed for detection of resistance mutations in EGFR-mutated NSCLC patients that may be linked with poor response on TKIs. Case Description Here, we present a case of an NSCLC patient harboring an EGFR somatic mutation and a concomitant leucine-zipper-like transcriptional regulator-1 (LZTR1) germline mutation. The patient experienced rapid disease progression on first-line EGFR TKI, osimertinib-chemotherapy, combination therapy with a PFS of only 4 months as compared to the median PFS of 27.9 months in the FLAURA2 study. Conclusions This case report indicates that identification of germline resistance mutations such as LZTR1 may be associated with poor response to EGFR TKIs. Furthermore, further characterization of these resistance mutations beyond somatic mutations can aid in development of future therapeutic options, which currently do not exist. It is recommended that germline testing be performed as part of the initial patient workup, if available.
Collapse
Affiliation(s)
- Wai Park
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Jeremy Fricke
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Isa Mambetsariev
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Giovanny Velasquez
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Reza Nadaf-Rahrov
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Shaira Therese Dingal
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Pauline Kim
- Department of Pharmacy, City of Hope, Duarte, CA, USA
| | - Razmig Babikian
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Arya Amini
- Department of Radiation Oncology, City of Hope, Duarte, CA, USA
| | | | - Ravi Salgia
- Department of Medical Oncology and Therapeutic Research, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
7
|
Li F, Yu Y, Jiang M, Zhang H. Targets for improving prostate tumor response to radiotherapy. Eur J Pharmacol 2025; 986:177149. [PMID: 39577551 DOI: 10.1016/j.ejphar.2024.177149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Prostate cancer is a prevalent malignancy that is frequently managed with radiotherapy. However, resistance to radiotherapy remains a significant challenge in controlling this disease. Early radiotherapy is employed for locally confined prostate cancer (PCa), while recurrent disease post-surgery and metastatic castration-resistant prostate cancer (mCRPC) are treated with late-stage radiotherapy, including radium-223. Combination therapies to integrate radiotherapy and chemotherapy have demonstrated enhanced treatment efficacy. Nonetheless, both modalities can induce severe local and systemic toxicities. Consequently, selectively sensitizing prostate tumors to radiotherapy could improve therapeutic outcomes while minimizing systemic side effects. The mechanisms underlying radioresistance in prostate cancer are multifaceted, including DNA damage repair (DDR) pathways, hypoxia, angiogenesis, androgen receptor (AR) signaling, and immune evasion. The advent of 177Lu-PSMA-617, which was approved in 2022, has shown promise in targeting prostate-specific membrane antigen (PSMA) in advanced prostate cancer. Experimental and clinical studies have yielded promising results in suppressing prostate tumors by targeting these pathways. This paper reviews potential targets for sensitizing prostate tumors to radiotherapy. We discuss cellular and molecular mechanisms contributing to therapy resistance and examine findings from experimental and clinical trials on promising targets and drugs that can be used in combination with radiotherapy.
Collapse
Affiliation(s)
- Fengguang Li
- Department of Urology, Yantaishan Hospital, Shandong, 264000, China
| | - Yizhi Yu
- Department of Urology, Yantaishan Hospital, Shandong, 264000, China
| | - Maozhu Jiang
- Department of Radiotherapy, Yantaishan Hospital, Shandong, 264000, China
| | - Haiying Zhang
- Department of Urology, Yantaishan Hospital, Shandong, 264000, China.
| |
Collapse
|
8
|
Obasi J, Sharma K, De Sarkar N, Antonarakis ES, Kilari D. Platinum Chemotherapy After PARP Inhibition in HRR-Deficient Metastatic Castration-Resistant Prostate Cancer. Clin Genitourin Cancer 2024; 22:102187. [PMID: 39241311 DOI: 10.1016/j.clgc.2024.102187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 09/09/2024]
Affiliation(s)
- Jennifer Obasi
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI; School of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Komal Sharma
- Medical College of Wisconsin Cancer Center, Milwaukee, WI; Department of Pathology, Medical College of Wisconsin, Milwaukee, WI; Data Science Institute, School of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Navonil De Sarkar
- Medical College of Wisconsin Cancer Center, Milwaukee, WI; Department of Pathology, Medical College of Wisconsin, Milwaukee, WI; Data Science Institute, School of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Emmanuel S Antonarakis
- Division of Hematology, Oncology, and Transplantation, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Deepak Kilari
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI.
| |
Collapse
|
9
|
Moosavi F, Hassani B, Nazari S, Saso L, Firuzi O. Targeting DNA damage response in pancreatic ductal adenocarcinoma: A review of preclinical and clinical evidence. Biochim Biophys Acta Rev Cancer 2024; 1879:189185. [PMID: 39326802 DOI: 10.1016/j.bbcan.2024.189185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with one of the most unfavorable prognoses across all malignancies. In this review, we investigate the role of inhibitors targeting crucial regulators of DNA damage response (DDR) pathways, either as single treatments or in combination with chemotherapeutic agents and targeted therapies in PDAC. The most prominent clinical benefit of PARP inhibitors' monotherapy is related to the principle of synthetic lethality in individuals harboring BRCA1/2 and other DDR gene mutations as predictive biomarkers. Moreover, induction of BRCAness with inhibitors of RTKs, including VEGFR and c-MET and their downstream signaling pathways, RAS/RAF/MEK/ERK and PI3K/AKT/mTOR in order to expand the application of PARP inhibitors in patients without DDR mutations, has also been addressed. Other DDR-targeting agents beyond PARP inhibitors, including inhibitors of ATM, ATR, CHEK1/2, and WEE1 have also demonstrated their potential in preclinical models of PDAC and may hold promise in future studies.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahareh Hassani
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Nazari
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
10
|
Miao R, Blue K, Sommerer K, Shah A, Bottiglieri S, del Cueto A, Berry DK, Ho TT, Hicks JK, Kim DW. PARP Inhibitors in Pancreatic Cancer with Homologous Recombination Repair Gene Mutations: A Single-Institution Experience. Cancers (Basel) 2024; 16:3447. [PMID: 39456541 PMCID: PMC11505755 DOI: 10.3390/cancers16203447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Limited data are available regarding the anticancer activity of PARP inhibitors (PARPis) in pancreatic cancer with mutations in HRR genes other than BRCA and PALB2. METHODS We retrospectively reviewed the clinical characteristics and outcomes of 48 patients with advanced pancreatic cancer harboring pathogenic germline and/or somatic HRR mutations who were treated with PARPis. RESULTS Thirty patients had germline (g)HRR mutations only, twelve had somatic (s)HRR mutations only, and six had concomitant gHRR and sHRR mutations. The objective response rate (ORR) was 22%. The median progression-free survival (mPFS) and overall survival (mOS) were 6.9 and 11.5 months, respectively. Five patients received olaparib in the front-line setting due to borderline performance status. Their ORR was 20%, and their mPFS and mOS were both 11.3 months. The ORR was higher in patients with BRCA or PALB2 mutations (germline or somatic) than in those with non-BRCA/PALB2 mutations. Patients with somatic non-BRCA/PALB2 variants had a shorter mPFS. Patients with concomitant gHRR/sHRR mutations or gHRR mutations alone had a significantly longer mPFS than those with sHRR mutations only. CONCLUSIONS PARP inhibitors may be considered for patients with advanced pancreatic cancer harboring pathogenic alterations of BRCA who cannot tolerate standard chemotherapy. Maintenance PARPis can be considered in selected patients with non-BRCA/non-PALB2 HRR mutations.
Collapse
Affiliation(s)
- Ruoyu Miao
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| | - Kirsten Blue
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| | - Katelyn Sommerer
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| | - Anand Shah
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| | - Sal Bottiglieri
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| | - Alex del Cueto
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA; (A.d.C.); (D.K.B.); (T.T.H.); (J.K.H.)
| | - Darcy K. Berry
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA; (A.d.C.); (D.K.B.); (T.T.H.); (J.K.H.)
| | - Teresa T. Ho
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA; (A.d.C.); (D.K.B.); (T.T.H.); (J.K.H.)
| | - James Kevin Hicks
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA; (A.d.C.); (D.K.B.); (T.T.H.); (J.K.H.)
| | - Dae Won Kim
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA; (R.M.); (K.B.); (K.S.); (A.S.); (S.B.)
| |
Collapse
|
11
|
Gaudio G, Martino E, Pellizzari G, Cavallone M, Castellano G, Omar A, Katselashvili L, Trapani D, Curigliano G. Developing combination therapies with biologics in triple-negative breast cancer. Expert Opin Biol Ther 2024; 24:1075-1094. [PMID: 39360776 DOI: 10.1080/14712598.2024.2408756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/22/2024] [Indexed: 10/11/2024]
Abstract
INTRODUCTION Novel compounds have entered the triple-negative breast cancer (TNBC) treatment algorithm, namely immune checkpoints inhibitors (ICIs), PARP inhibitors and antibody-drug conjugates (ADCs). The optimization of treatment efficacy can be enhanced with the use of combination treatments, and the incorporation of novel compounds. In this review, we discuss the combination treatments under development for the treatment of TNBC. AREAS COVERED The development of new drugs occurring in recent years has boosted the research for novel combinations to target TNBC heterogeneity and improve outcomes. ICIs, ADCs, tyrosine kinase inhibitors (TKIs), and PARP inhibitors have emerged as leading players in this new landscape, while other compounds like novel intracellular pathways inhibitors or cancer vaccines are drawing more and more interest. The future of TNBC is outlined in combination approaches, and based on new cancer targets, including many chemotherapy-free treatments. EXPERT OPINION A large number of TNBC therapies have either proved clinically ineffective or weighted by unacceptable safety profiles. Others, however, have provided promising results and are currently in late-stage clinical trials, while a few have actually changed clinical practice in recent years. As novel, more and more selective drugs come up, combination strategies focusing the concept of synergy are fully warranted for the future.
Collapse
Affiliation(s)
- Gilda Gaudio
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Enzo Martino
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Gloria Pellizzari
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Matteo Cavallone
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Grazia Castellano
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Abeid Omar
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Nuclear Medicine, Kenyatta University Teaching Referral and Research Hospital, Nairobi, Kenya
| | - Lika Katselashvili
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology, Caucasus Medical Centre, Tbilisi, Georgia
| | - Dario Trapani
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology (DIPO), University of Milan, Milan, Italy
| |
Collapse
|
12
|
Ilié M, Heeke S, Horgan D, Hofman P. Navigating Change in Tumor Naming: Exploring the Complexities and Considerations of Shifting Toward Molecular Classifications. J Clin Oncol 2024; 42:3183-3186. [PMID: 38935877 DOI: 10.1200/jco.24.00323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/21/2024] [Accepted: 05/08/2024] [Indexed: 06/29/2024] Open
Abstract
Navigating change in tumor naming. Balance organ-based and molecular classifications for optimal treatment.
Collapse
Affiliation(s)
- Marius Ilié
- Laboratory of Clinical and Experimental Pathology, Hospital-Integrated Biobank (BB-0033-00025), IHU RespirERA, FHU OncoAge, University Hospital Centre Nice, University Côte d'Azur, Nice, France
| | - Simon Heeke
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Denis Horgan
- European Alliance for Personalised Medicine, Brussels, Belgium
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Hospital-Integrated Biobank (BB-0033-00025), IHU RespirERA, FHU OncoAge, University Hospital Centre Nice, University Côte d'Azur, Nice, France
| |
Collapse
|
13
|
Hayman TJ. Rethinking the use of germline CHEK2 mutation as a marker for PARP inhibitor sensitivity. JNCI Cancer Spectr 2024; 8:pkae045. [PMID: 38950525 PMCID: PMC11216723 DOI: 10.1093/jncics/pkae045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 06/02/2024] [Indexed: 07/03/2024] Open
Affiliation(s)
- Thomas J Hayman
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
14
|
El-Sayed MM, Bianco JR, Li Y, Fabian Z. Tumor-Agnostic Therapy-The Final Step Forward in the Cure for Human Neoplasms? Cells 2024; 13:1071. [PMID: 38920700 PMCID: PMC11201516 DOI: 10.3390/cells13121071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
Cancer accounted for 10 million deaths in 2020, nearly one in every six deaths annually. Despite advancements, the contemporary clinical management of human neoplasms faces a number of challenges. Surgical removal of tumor tissues is often not possible technically, while radiation and chemotherapy pose the risk of damaging healthy cells, tissues, and organs, presenting complex clinical challenges. These require a paradigm shift in developing new therapeutic modalities moving towards a more personalized and targeted approach. The tumor-agnostic philosophy, one of these new modalities, focuses on characteristic molecular signatures of transformed cells independently of their traditional histopathological classification. These include commonly occurring DNA aberrations in cancer cells, shared metabolic features of their homeostasis or immune evasion measures of the tumor tissues. The first dedicated, FDA-approved tumor-agnostic agent's profound progression-free survival of 78% in mismatch repair-deficient colorectal cancer paved the way for the accelerated FDA approvals of novel tumor-agnostic therapeutic compounds. Here, we review the historical background, current status, and future perspectives of this new era of clinical oncology.
Collapse
Affiliation(s)
| | | | | | - Zsolt Fabian
- School of Medicine and Dentistry, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK; (M.M.E.-S.); (J.R.B.); (Y.L.)
| |
Collapse
|
15
|
Carsote M, Nistor C, Gheorghe AM, Sima OC, Trandafir AI, Nistor TVI, Sandulescu BA, Ciobica ML. Turning Points in Cross-Disciplinary Perspective of Primary Hyperparathyroidism and Pancreas Involvements: Hypercalcemia-Induced Pancreatitis, MEN1 Gene-Related Tumors, and Insulin Resistance. Int J Mol Sci 2024; 25:6349. [PMID: 38928056 PMCID: PMC11203827 DOI: 10.3390/ijms25126349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/13/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
We aimed to provide an in-depth analysis with respect to three turning points in pancreas involvement in primary hyperparathyroidism (PHP): hypercalcemia-induced pancreatitis (HCa-P), MEN1 (multiple endocrine neoplasia)-related neuroendocrine tumors (NETs), and insulin resistance (IR). This was a comprehensive review conducted via a PubMed search between January 2020 and January 2024. HCa-P (n = 9 studies, N = 1375) involved as a starting point parathyroid NETs (n = 7) or pancreatitis (n = 2, N = 167). Case report-focused analysis (N = 27) showed five cases of pregnancy PHP-HCa-P and three reports of parathyroid carcinoma (female/male ratio of 2/1, ages of 34 in women, men of 56). MEN1-NET studies (n = 7) included MEN1-related insulinomas (n = 2) or MEN1-associated PHP (n = 2) or analyses of genetic profile (n = 3), for a total of 877 MEN1 subjects. In MEN1 insulinomas (N = 77), the rate of associated PHP was 78%. Recurrence after parathyroidectomy (N = 585 with PHP) was higher after less-than-subtotal versus subtotal parathyroidectomy (68% versus 45%, p < 0.001); re-do surgery was 26% depending on surgery for pancreatic NETs (found in 82% of PHP patients). MEN1 pathogenic variants in exon 10 represented an independent risk factor for PHP recurrence. A single pediatric study in MEN1 (N = 80) revealed the following: a PHP rate of 80% and pancreatic NET rate of 35% and 35 underlying germline MEN1 pathogenic variants (and 3/35 of them were newly detected). The co-occurrence of genetic anomalies included the following: CDC73 gene variant, glucokinase regulatory protein gene pathogenic variant (c.151C>T, p.Arg51*), and CAH-X syndrome. IR/metabolic feature-focused analysis identified (n = 10, N = 1010) a heterogeneous spectrum: approximately one-third of adults might have had prediabetes, almost half displayed some level of IR as reflected by HOMA-IR > 2.6, and serum calcium was positively correlated with HOMA-IR. Vitamin D deficiency was associated with a higher rate of metabolic syndrome (n = 1). Normocalcemic and mildly symptomatic hyperparathyroidism (n = 6, N = 193) was associated with a higher fasting glucose and some improvement after parathyroidectomy. This multilayer pancreas/parathyroid analysis highlighted a complex panel of connections from pathogenic factors, including biochemical, molecular, genetic, and metabolic factors, to a clinical multidisciplinary panel.
Collapse
Affiliation(s)
- Mara Carsote
- Department of Endocrinology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Clinical Endocrinology V, “C.I. Parhon” National Institute of Endocrinology, 011863 Bucharest, Romania
| | - Claudiu Nistor
- Department 4-Cardio-Thoracic Pathology, Thoracic Surgery II Discipline, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Thoracic Surgery Department, “Dr. Carol Davila” Central Military University Emergency Hospital, 010242 Bucharest, Romania
| | - Ana-Maria Gheorghe
- PhD Doctoral School, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-M.G.); (O.-C.S.); (A.-I.T.); (B.-A.S.)
| | - Oana-Claudia Sima
- PhD Doctoral School, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-M.G.); (O.-C.S.); (A.-I.T.); (B.-A.S.)
| | - Alexandra-Ioana Trandafir
- PhD Doctoral School, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-M.G.); (O.-C.S.); (A.-I.T.); (B.-A.S.)
| | - Tiberiu Vasile Ioan Nistor
- Department of Clinical Biochemistry, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Bianca-Andreea Sandulescu
- PhD Doctoral School, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-M.G.); (O.-C.S.); (A.-I.T.); (B.-A.S.)
- Department of Internal Medicine and Gastroenterology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Internal Medicine I and Rheumatology, “Dr. Carol Davila” Central Military University Emergency Hospital, 010825 Bucharest, Romania
| | - Mihai-Lucian Ciobica
- Department of Internal Medicine and Gastroenterology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Internal Medicine I and Rheumatology, “Dr. Carol Davila” Central Military University Emergency Hospital, 010825 Bucharest, Romania
| |
Collapse
|
16
|
Safyan RA, Kim E, Dekker E, Homs M, Aguirre AJ, Koerkamp BG, Chiorean EG. Multidisciplinary Standards and Evolving Therapies for Patients With Pancreatic Cancer. Am Soc Clin Oncol Educ Book 2024; 44:e438598. [PMID: 38781541 DOI: 10.1200/edbk_438598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a challenging disease that presents at an advanced stage and results in many symptoms that negatively influence patients' quality of life and reduce their ability to receive effective treatment. Early implementation of expert multidisciplinary care with nutritional support, exercise, and palliative care for both early-stage and advanced disease promises to maintain or improve the patients' physical, social, and psychological well-being, decrease aggressive interventions at the end of life, and ultimately improve survival. Moreover, advances in treatment strategies in the neoadjuvant and metastatic setting combined with novel therapeutic agents targeting the key drivers of the disease are leading to improvements in the care of patients with pancreatic cancer. Here, we emphasize the multidisciplinary supportive and therapeutic care of patients with PDA, review current guidelines and new developments of neoadjuvant and perioperative treatments for localized disease, as well as the treatment standards and the evolving field of precision oncology and immunotherapies for advanced PDA.
Collapse
Affiliation(s)
- Rachael A Safyan
- University of Washington School of Medicine, Department of Medicine, Division of Hematology-Oncology, Seattle, WA
- Fred Hutchinson Cancer Center, Clinical Research Division, Seattle, WA
| | - Eejung Kim
- Dana-Farber Cancer Center, Department of Medical Oncology, Boston, MA
- Harvard Medical School, Boston, MA
| | - Emmelie Dekker
- Erasmus MC Cancer Institute, Department of Surgery, Rotterdam, the Netherlands
| | - Marjolein Homs
- Erasmus MC Cancer Institute, Department of Medical Oncology, Rotterdam, the Netherlands
| | - Andrew J Aguirre
- Dana-Farber Cancer Center, Department of Medical Oncology, Boston, MA
- Harvard Medical School, Boston, MA
| | - Bas Groot Koerkamp
- Erasmus MC Cancer Institute, Department of Surgery, Rotterdam, the Netherlands
| | - E Gabriela Chiorean
- University of Washington School of Medicine, Department of Medicine, Division of Hematology-Oncology, Seattle, WA
- Fred Hutchinson Cancer Center, Clinical Research Division, Seattle, WA
| |
Collapse
|
17
|
Harrold EC, Stadler ZK. Upper Gastrointestinal Cancers and the Role of Genetic Testing. Hematol Oncol Clin North Am 2024; 38:677-691. [PMID: 38458854 DOI: 10.1016/j.hoc.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2024]
Abstract
Beyond the few established hereditary cancer syndromes with an upper gastrointestinal cancer component, there is increasing recognition of the contribution of novel pathogenic germline variants (gPVs) to upper gastrointestinal carcinogenesis. The detection of gPVs has potential implications for novel treatment approaches of the index cancer patient as well as long-term implications for surveillance and risk-reducing measures for cancer survivors and far-reaching implications for the patients' family. With widespread availability of multigene panel testing, new associations may be identified with germline-somatic integration being critical to determining true causality of novel gPVs. Comprehensive cancer care should incorporate both somatic and germline testing.
Collapse
Affiliation(s)
- Emily C Harrold
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medical Oncology, Mater Misericordiae University Hospital, Dublin, Ireland. https://twitter.com/EmilyHarrold6
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
18
|
Aleksakhina SN, Ivantsov AO, Imyanitov EN. Agnostic Administration of Targeted Anticancer Drugs: Looking for a Balance between Hype and Caution. Int J Mol Sci 2024; 25:4094. [PMID: 38612902 PMCID: PMC11012409 DOI: 10.3390/ijms25074094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024] Open
Abstract
Many tumors have well-defined vulnerabilities, thus potentially allowing highly specific and effective treatment. There is a spectrum of actionable genetic alterations which are shared across various tumor types and, therefore, can be targeted by a given drug irrespective of tumor histology. Several agnostic drug-target matches have already been approved for clinical use, e.g., immune therapy for tumors with microsatellite instability (MSI) and/or high tumor mutation burden (TMB), NTRK1-3 and RET inhibitors for cancers carrying rearrangements in these kinases, and dabrafenib plus trametinib for BRAF V600E mutated malignancies. Multiple lines of evidence suggest that this histology-independent approach is also reasonable for tumors carrying ALK and ROS1 translocations, biallelic BRCA1/2 inactivation and/or homologous recombination deficiency (HRD), strong HER2 amplification/overexpression coupled with the absence of other MAPK pathway-activating mutations, etc. On the other hand, some well-known targets are not agnostic: for example, PD-L1 expression is predictive for the efficacy of PD-L1/PD1 inhibitors only in some but not all cancer types. Unfortunately, the individual probability of finding a druggable target in a given tumor is relatively low, even with the use of comprehensive next-generation sequencing (NGS) assays. Nevertheless, the rapidly growing utilization of NGS will significantly increase the number of patients with highly unusual or exceptionally rare tumor-target combinations. Clinical trials may provide only a framework for treatment attitudes, while the decisions for individual patients usually require case-by-case consideration of the probability of deriving benefit from agnostic versus standard therapy, drug availability, associated costs, and other circumstances. The existing format of data dissemination may not be optimal for agnostic cancer medicine, as conventional scientific journals are understandably biased towards the publication of positive findings and usually discourage the submission of case reports. Despite all the limitations and concerns, histology-independent drug-target matching is certainly feasible and, therefore, will be increasingly utilized in the future.
Collapse
Affiliation(s)
- Svetlana N. Aleksakhina
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
| | - Alexander O. Ivantsov
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| | - Evgeny N. Imyanitov
- Department of Tumor Growth Biology, N. N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia
- Department of Medical Genetics, St. Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| |
Collapse
|