Pub Date : 2024-03-12DOI: 10.1016/j.ctrv.2024.102719
Elena Brozos-Vázquez , Marta Toledano-Fonseca , Nicolás Costa-Fraga , María Victoria García-Ortiz , Ángel Díaz-Lagares , Antonio Rodríguez-Ariza , Enrique Aranda , Rafael López-López
Pancreatic cancer is one of the tumors with the worst prognosis, and unlike other cancers, few advances have been made in recent years. The only curative option is surgery, but only 15–20% of patients are candidates, with a high risk of relapse. In advanced pancreatic cancer there are few first-line treatment options and no validated biomarkers for better treatment selection. The development of targeted therapies in pancreatic cancer is increasingly feasible due to tumor-agnostic treatments, such as PARP inhibitors in patients with BRCA1, BRCA2 or PALB2 alterations or immunotherapies in patients with high microsatellite instability/tumor mutational burden. In addition, other therapeutic molecules have been developed for patients with KRAS G12C mutation or fusions in NTRK or NRG1. Consequently, there has been a growing interest in biomarkers that may help guide targeted therapy in pancreatic cancer. Therefore, this review aims to offer an updated perspective on biomarkers with therapeutic potential in pancreatic cancer.
{"title":"Pancreatic cancer biomarkers: A pathway to advance in personalized treatment selection","authors":"Elena Brozos-Vázquez , Marta Toledano-Fonseca , Nicolás Costa-Fraga , María Victoria García-Ortiz , Ángel Díaz-Lagares , Antonio Rodríguez-Ariza , Enrique Aranda , Rafael López-López","doi":"10.1016/j.ctrv.2024.102719","DOIUrl":"https://doi.org/10.1016/j.ctrv.2024.102719","url":null,"abstract":"<div><p>Pancreatic cancer is one of the tumors with the worst prognosis, and unlike other cancers, few advances have been made in recent years. The only curative option is surgery, but only 15–20% of patients are candidates, with a high risk of relapse. In advanced pancreatic cancer there are few first-line treatment options and no validated biomarkers for better treatment selection. The development of targeted therapies in pancreatic cancer is increasingly feasible due to tumor-agnostic treatments, such as PARP inhibitors in patients with <em>BRCA1</em>, <em>BRCA2</em> or <em>PALB2</em> alterations or immunotherapies in patients with high microsatellite instability/tumor mutational burden. In addition, other therapeutic molecules have been developed for patients with <em>KRAS</em> G12C mutation or fusions in <em>NTRK</em> or <em>NRG1</em>. Consequently, there has been a growing interest in biomarkers that may help guide targeted therapy in pancreatic cancer. Therefore, this review aims to offer an updated perspective on biomarkers with therapeutic potential in pancreatic cancer.</p></div>","PeriodicalId":9537,"journal":{"name":"Cancer treatment reviews","volume":"125 ","pages":"Article 102719"},"PeriodicalIF":11.8,"publicationDate":"2024-03-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S030573722400046X/pdfft?md5=96ec3537f15005b18419db44251fc740&pid=1-s2.0-S030573722400046X-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140122558","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-03-11DOI: 10.1016/j.ctrv.2024.102716
A. Kyriazoglou , A. Pagkali , I. Kotsantis , P. Economopoulou , M. Kyrkasiadou , M. Moutafi , N. Gavrielatou , M. Anastasiou , A. Boulouta , A. Pantazopoulos , M. Giannakakou , A. Digklia , A. Psyrri
Well-differentiated liposarcomas (WDLPS) and dedifferentiated liposarcomas (DDLPS) account for 60 % of all liposarcomas, reflecting the heterogeneity of this type of sarcoma. Genetically, both types of liposarcomas are characterized by the amplification of MDM2 and CDK4 genes, which indicates an important molecular event with diagnostic and therapeutic relevance. In both localized WDLPS and DDLPS of the retroperitoneum and the extremities, between 25 % and 30 % of patients have local or distant recurrence, even when perioperatively treated, with clear margins present. The systemic treatment of WDLPS and DDLPS remains a challenge, with anthracyclines as the gold standard for first-line treatment. Several regimens have been tested with modest results regarding their efficacy. Herein we discuss the systemic treatment options for WDLPS and DDLPS and review their reported clinical efficacy results.
{"title":"Well-differentiated liposarcomas and dedifferentiated liposarcomas: Systemic treatment options for two sibling neoplasms","authors":"A. Kyriazoglou , A. Pagkali , I. Kotsantis , P. Economopoulou , M. Kyrkasiadou , M. Moutafi , N. Gavrielatou , M. Anastasiou , A. Boulouta , A. Pantazopoulos , M. Giannakakou , A. Digklia , A. Psyrri","doi":"10.1016/j.ctrv.2024.102716","DOIUrl":"10.1016/j.ctrv.2024.102716","url":null,"abstract":"<div><p>Well-differentiated liposarcomas (WDLPS) and dedifferentiated liposarcomas (DDLPS) account for 60 % of all liposarcomas, reflecting the heterogeneity of this type of sarcoma. Genetically, both types of liposarcomas are characterized by the amplification of <em>MDM2</em> and <em>CDK4</em> genes, which indicates an important molecular event with diagnostic and therapeutic relevance. In both localized WDLPS and DDLPS of the retroperitoneum and the extremities, between 25 % and 30 % of patients have local or distant recurrence, even when perioperatively treated, with clear margins present. The systemic treatment of WDLPS and DDLPS remains a challenge, with anthracyclines as the gold standard for first-line treatment. Several regimens have been tested with modest results regarding their efficacy. Herein we discuss the systemic treatment options for WDLPS and DDLPS and review their reported clinical efficacy results.</p></div>","PeriodicalId":9537,"journal":{"name":"Cancer treatment reviews","volume":"125 ","pages":"Article 102716"},"PeriodicalIF":11.8,"publicationDate":"2024-03-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140124695","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-03-11DOI: 10.1016/j.ctrv.2024.102720
Rebecca S. Heist , Jacob Sands , Aditya Bardia , Toshio Shimizu , Aaron Lisberg , Ian Krop , Noboru Yamamoto , Takahiro Kogawa , Saba Al-Hashimi , Simon S.M. Fung , Anat Galor , Francesca Pisetzky , Priyanka Basak , Cindy Lau , Funda Meric-Bernstam
Antibody drug conjugates (ADCs) are an emerging class of treatments designed to improve efficacy and decrease toxicity compared with other systemic therapies through the selective delivery of cytotoxic agents to tumor cells. Datopotamab deruxtecan (Dato-DXd) is a novel ADC comprising a topoisomerase I inhibitor payload and a monoclonal antibody directed to trophoblast cell-surface antigen 2 (TROP2), a protein that is broadly expressed in several types of solid tumors. Dato-DXd is being investigated across multiple solid tumor indications. In the ongoing, first-in-human TROPION-PanTumor01 phase I study (ClinicalTrials.gov: NCT03401385), encouraging and durable antitumor activity and a manageable safety profile was demonstrated in patients with advanced/metastatic hormone receptor-positive/human epidermal growth factor receptor 2-negative breast cancer (HR+/HER2– BC), triple-negative breast cancer (TNBC), and non-small cell lung cancer (NSCLC).
Improved understanding of the adverse events (AEs) that are associated with Dato-DXd and their optimal management is essential to ensure safe and successful administration. Interstitial lung disease/pneumonitis, infusion-related reactions, oral mucositis/stomatitis, and ocular surface events have been identified as AEs of special interest (AESIs) for which appropriate prevention, monitoring, and management is essential. This article summarizes the incidence of AESIs among patients with HR+/HER2− BC, TNBC, and NSCLC reported in TROPION-PanTumor01. We report our recommendations for AESI prophylaxis, early detection, and management, using experience gained from treating AESIs that occur with Dato-DXd in clinical trials.
抗体药物共轭物(ADC)是一种新兴的治疗方法,与其他全身治疗方法相比,它通过选择性地向肿瘤细胞输送细胞毒性药物来提高疗效和降低毒性。Datopotamab deruxtecan(Dato-DXd)是一种新型ADC,由拓扑异构酶I抑制剂有效载荷和针对滋养层母细胞表面抗原2(TROP2)的单克隆抗体组成。Dato-DXd 正在多个实体瘤适应症中进行研究。在正在进行的首个人体 TROPION-PanTumor01 I 期研究(ClinicalTrials.gov:NCT03401385)中,激素受体阳性/人类表皮生长因子受体2阴性的晚期/转移性乳腺癌(HR+/HER2- BC)、三阴性乳腺癌(TNBC)和非小细胞肺癌(NSCLC)患者表现出令人鼓舞的持久抗肿瘤活性和可控的安全性。
{"title":"Clinical management, monitoring, and prophylaxis of adverse events of special interest associated with datopotamab deruxtecan","authors":"Rebecca S. Heist , Jacob Sands , Aditya Bardia , Toshio Shimizu , Aaron Lisberg , Ian Krop , Noboru Yamamoto , Takahiro Kogawa , Saba Al-Hashimi , Simon S.M. Fung , Anat Galor , Francesca Pisetzky , Priyanka Basak , Cindy Lau , Funda Meric-Bernstam","doi":"10.1016/j.ctrv.2024.102720","DOIUrl":"10.1016/j.ctrv.2024.102720","url":null,"abstract":"<div><p>Antibody drug conjugates (ADCs) are an emerging class of treatments designed to improve efficacy and decrease toxicity compared with other systemic therapies through the selective delivery of cytotoxic agents to tumor cells. Datopotamab deruxtecan (Dato-DXd) is a novel ADC comprising a topoisomerase I inhibitor payload and a monoclonal antibody directed to trophoblast cell-surface antigen 2 (TROP2), a protein that is broadly expressed in several types of solid tumors. Dato-DXd is being investigated across multiple solid tumor indications. In the ongoing, first-in-human TROPION-PanTumor01 phase I study (ClinicalTrials.gov: NCT03401385), encouraging and durable antitumor activity and a manageable safety profile was demonstrated in patients with advanced/metastatic hormone receptor-positive/human epidermal growth factor receptor<!--> <!-->2-negative breast cancer (HR+/HER2– BC), triple-negative breast cancer (TNBC), and non-small cell lung cancer (NSCLC).</p><p>Improved understanding of the adverse events (AEs) that are associated with Dato-DXd and their optimal management is essential to ensure safe and successful administration. Interstitial lung disease/pneumonitis, infusion-related reactions, oral mucositis/stomatitis, and ocular surface events have been identified as AEs of special interest (AESIs) for which appropriate prevention, monitoring, and management is essential. This article summarizes the incidence of AESIs among patients with HR+/HER2− BC, TNBC, and NSCLC reported in TROPION-PanTumor01. We report our recommendations for AESI prophylaxis, early detection, and management, using experience gained from treating AESIs that occur with Dato-DXd in clinical trials.</p></div>","PeriodicalId":9537,"journal":{"name":"Cancer treatment reviews","volume":"125 ","pages":"Article 102720"},"PeriodicalIF":11.8,"publicationDate":"2024-03-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0305737224000471/pdfft?md5=42e58372bfd4d21d42fccffff25e1a90&pid=1-s2.0-S0305737224000471-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140124591","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-03-05DOI: 10.1016/j.ctrv.2024.102715
Alexander Grunenberg , Christian Buske
Clinical management of Waldenström’s Macroglobulinemia has seen major progress in the recent years, triggered by our improved understanding of the biology of the disease and the development of new therapies. Based on this there are multiple treatment options available for patients with WM ranging from classical immunochemotherapy to targeted approaches blocking key enzymes involved in lymphoma growth. This review summarizes our current knowledge about diagnostics and treatment of this rare but recurrent lymphoma subtype, which often presents a major clinical challenge in daily clinical life.
{"title":"How to manage waldenström’s macroglobulinemia in 2024","authors":"Alexander Grunenberg , Christian Buske","doi":"10.1016/j.ctrv.2024.102715","DOIUrl":"10.1016/j.ctrv.2024.102715","url":null,"abstract":"<div><p>Clinical management of Waldenström’s Macroglobulinemia has seen major progress in the recent years, triggered by our improved understanding of the biology of the disease and the development of new therapies. Based on this there are multiple treatment options available for patients with WM ranging from classical immunochemotherapy to targeted approaches blocking key enzymes involved in lymphoma growth. This review summarizes our current knowledge about diagnostics and treatment of this rare but recurrent lymphoma subtype, which often presents a major clinical challenge in daily clinical life.</p></div>","PeriodicalId":9537,"journal":{"name":"Cancer treatment reviews","volume":"125 ","pages":"Article 102715"},"PeriodicalIF":11.8,"publicationDate":"2024-03-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140055896","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-03-04DOI: 10.1016/j.ctrv.2024.102703
Elena Fountzilas , Apostolia-Maria Tsimberidou , Henry Hiep Vo , Razelle Kurzrock
Choosing the right drug(s) for the right patient via advanced genomic sequencing and multi-omic interrogation is the sine qua non of precision cancer medicine. Traditional cancer clinical trial designs follow well-defined protocols to evaluate the efficacy of new therapies in patient groups, usually identified by their histology/tissue of origin of their malignancy. In contrast, precision medicine seeks to optimize benefit in individual patients, i.e., to define who benefits rather than determine whether the overall group benefits. Since cancer is a disease driven by molecular alterations, innovative trial designs, including biomarker-defined tumor-agnostic basket trials, are driving ground-breaking regulatory approvals and deployment of gene- and immune-targeted drugs. Molecular interrogation further reveals the disruptive reality that advanced cancers are extraordinarily complex and individually distinct. Therefore, optimized treatment often requires drug combinations and N-of-1 customization, addressed by a new generation of N-of-1 trials. Real-world data and structured master registry trials are also providing massive datasets that are further fueling a transformation in oncology. Finally, machine learning is facilitating rapid discovery, and it is plausible that high-throughput computing, in silico modeling, and 3-dimensional printing may be exploitable in the near future to discover and design customized drugs in real time.
{"title":"Tumor-agnostic baskets to N-of-1 platform trials and real-world data: Transforming precision oncology clinical trial design","authors":"Elena Fountzilas , Apostolia-Maria Tsimberidou , Henry Hiep Vo , Razelle Kurzrock","doi":"10.1016/j.ctrv.2024.102703","DOIUrl":"10.1016/j.ctrv.2024.102703","url":null,"abstract":"<div><p>Choosing the right drug(s) for the right patient via advanced genomic sequencing and multi-omic interrogation is the <em>sine qua non</em> of precision cancer medicine. Traditional cancer clinical trial designs follow well-defined protocols to evaluate the efficacy of new therapies in patient groups, usually identified by their histology/tissue of origin of their malignancy. In contrast, precision medicine seeks to optimize benefit in individual patients, i.e., to define who benefits rather than determine whether the overall group benefits. Since cancer is a disease driven by molecular alterations, innovative trial designs, including biomarker-defined tumor-agnostic basket trials, are driving ground-breaking regulatory approvals and deployment of gene- and immune-targeted drugs. Molecular interrogation further reveals the disruptive reality that advanced cancers are extraordinarily complex and individually distinct. Therefore, optimized treatment often requires drug combinations and N-of-1 customization, addressed by a new generation of N-of-1 trials. Real-world data and structured master registry trials are also providing massive datasets that are further fueling a transformation in oncology. Finally, machine learning is facilitating rapid discovery, and it is plausible that high-throughput computing, <em>in silico</em> modeling, and 3-dimensional printing may be exploitable in the near future to discover and design customized drugs in real time.</p></div>","PeriodicalId":9537,"journal":{"name":"Cancer treatment reviews","volume":"125 ","pages":"Article 102703"},"PeriodicalIF":11.8,"publicationDate":"2024-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140055973","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-03-01DOI: 10.1016/j.ctrv.2024.102704
K.R.J. Kistemaker , F. Sijani , D.J. Brinkman , A. de Graeff , G.L. Burchell , M.A.H. Steegers , L. van Zuylen
<div><h3>Background</h3><p>Cancer-related pain often requires opioid treatment with opioid-induced constipation (OIC) as its most frequent gastrointestinal side-effect. Both for prevention and treatment of OIC osmotic (e.g. polyethylene glycol) and stimulant (e.g. bisacodyl) laxatives are widely used. Newer drugs such as the peripherally acting µ-opioid receptor antagonists (PAMORAs) and naloxone in a fixed combination with oxycodone have become available for the management of OIC.</p><p>This systematic review and meta-analysis aims to give an overview of the scientific evidence on pharmacological strategies for the prevention and treatment of OIC in cancer patients.</p></div><div><h3>Methods</h3><p>A systematic search in PubMed, Embase, Web of Science and the Cochrane Library was completed from inception up to 22 October 2022. Randomized and non-randomized studies were systematically selected. Bowel function and adverse drug events were assessed.</p></div><div><h3>Results</h3><p>Twenty trials (prevention: five RCTs and three cohort studies; treatment: ten RCTs and two comparative cohort studies) were included in the review.</p><p>Regarding the prevention of OIC, three RCTs compared laxatives with other laxatives, finding no clear differences in effectivity of the laxatives used. One cohort study showed a significant benefit of magnesium oxide compared with no laxative. One RCT found a significant benefit for the PAMORA naldemedine compared with magnesium oxide. Preventive use of oxycodone/naloxone did not show a significant difference in two out of three other studies compared to oxycodone or fentanyl. A meta-analysis was not possible.</p><p>Regarding the treatment of OIC, two RCTs compared laxatives, of which one RCT found that polyethylene glycol was significantly more effective than sennosides. Seven studies compared an opioid antagonist (naloxone, methylnaltrexone or naldemedine) with placebo and three studies compared different dosages of opioid antagonists. These studies with opioid antagonists were used for the meta-analysis.</p><p>Oxycodone/naloxone showed a significant improvement in Bowel Function Index compared to oxycodone with laxatives (MD −13.68; 95 % CI −18.38 to −8.98; I<sup>2</sup> = 58 %). Adverse drug event rates were similar amongst both groups, except for nausea in favour of oxycodone/naloxone (RR 0.51; 95 % CI 0.31–0.83; I<sup>2</sup> = 0 %). Naldemedine (NAL) and methylnaltrexone (MNTX) demonstrated significantly higher response rates compared to placebo (NAL: RR 2.07, 95 % CI 1.64–2.61, I<sup>2</sup> = 0 %; MNTX: RR 3.83, 95 % CI 2.81–5.22, I<sup>2</sup> = 0 %). With regard to adverse events, abdominal pain was more present in treatment with methylnaltrexone and diarrhea was significantly more present in treatment with naldemedine. Different dosages of methylnaltrexone were not significantly different with regard to both efficacy and adverse drug event rates.</p></div><div><h3>Conclusions</h3><p>Magnesium oxide and nalde
{"title":"Pharmacological prevention and treatment of opioid-induced constipation in cancer patients: A systematic review and meta-analysis","authors":"K.R.J. Kistemaker , F. Sijani , D.J. Brinkman , A. de Graeff , G.L. Burchell , M.A.H. Steegers , L. van Zuylen","doi":"10.1016/j.ctrv.2024.102704","DOIUrl":"https://doi.org/10.1016/j.ctrv.2024.102704","url":null,"abstract":"<div><h3>Background</h3><p>Cancer-related pain often requires opioid treatment with opioid-induced constipation (OIC) as its most frequent gastrointestinal side-effect. Both for prevention and treatment of OIC osmotic (e.g. polyethylene glycol) and stimulant (e.g. bisacodyl) laxatives are widely used. Newer drugs such as the peripherally acting µ-opioid receptor antagonists (PAMORAs) and naloxone in a fixed combination with oxycodone have become available for the management of OIC.</p><p>This systematic review and meta-analysis aims to give an overview of the scientific evidence on pharmacological strategies for the prevention and treatment of OIC in cancer patients.</p></div><div><h3>Methods</h3><p>A systematic search in PubMed, Embase, Web of Science and the Cochrane Library was completed from inception up to 22 October 2022. Randomized and non-randomized studies were systematically selected. Bowel function and adverse drug events were assessed.</p></div><div><h3>Results</h3><p>Twenty trials (prevention: five RCTs and three cohort studies; treatment: ten RCTs and two comparative cohort studies) were included in the review.</p><p>Regarding the prevention of OIC, three RCTs compared laxatives with other laxatives, finding no clear differences in effectivity of the laxatives used. One cohort study showed a significant benefit of magnesium oxide compared with no laxative. One RCT found a significant benefit for the PAMORA naldemedine compared with magnesium oxide. Preventive use of oxycodone/naloxone did not show a significant difference in two out of three other studies compared to oxycodone or fentanyl. A meta-analysis was not possible.</p><p>Regarding the treatment of OIC, two RCTs compared laxatives, of which one RCT found that polyethylene glycol was significantly more effective than sennosides. Seven studies compared an opioid antagonist (naloxone, methylnaltrexone or naldemedine) with placebo and three studies compared different dosages of opioid antagonists. These studies with opioid antagonists were used for the meta-analysis.</p><p>Oxycodone/naloxone showed a significant improvement in Bowel Function Index compared to oxycodone with laxatives (MD −13.68; 95 % CI −18.38 to −8.98; I<sup>2</sup> = 58 %). Adverse drug event rates were similar amongst both groups, except for nausea in favour of oxycodone/naloxone (RR 0.51; 95 % CI 0.31–0.83; I<sup>2</sup> = 0 %). Naldemedine (NAL) and methylnaltrexone (MNTX) demonstrated significantly higher response rates compared to placebo (NAL: RR 2.07, 95 % CI 1.64–2.61, I<sup>2</sup> = 0 %; MNTX: RR 3.83, 95 % CI 2.81–5.22, I<sup>2</sup> = 0 %). With regard to adverse events, abdominal pain was more present in treatment with methylnaltrexone and diarrhea was significantly more present in treatment with naldemedine. Different dosages of methylnaltrexone were not significantly different with regard to both efficacy and adverse drug event rates.</p></div><div><h3>Conclusions</h3><p>Magnesium oxide and nalde","PeriodicalId":9537,"journal":{"name":"Cancer treatment reviews","volume":"125 ","pages":"Article 102704"},"PeriodicalIF":11.8,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0305737224000227/pdfft?md5=71786d5672fa6b4f1ee184e40fb33ac1&pid=1-s2.0-S0305737224000227-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140041820","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-02-28DOI: 10.1016/j.ctrv.2024.102702
M. Pensabene , A. Calabrese , C. von Arx, R. Caputo, M. De Laurentiis
A relevant percentage of breast cancers (BCs) are tied to pathogenetic (P)/likely pathogenetic (LP) variants in predisposing genes. The knowledge of P/LP variants is an essential element in the management of BC patients since the first diagnosis because it influences surgery and subsequent oncological treatments and follow-up. Moreover, patients with metastatic BCs can benefit from personalized treatment if carriers of P/LP in BRCA1/2 genes. Multigene panels allow the identification of other predisposing genes with an impact on management. Cascade genetic testing for healthy family members allows personalized preventive strategies. Here, we review the advances and the challenges of Cancer Genetic Counseling (CGC). We focus on the area of oncology directed to hereditary BC management describing the peculiar way to lead CGC and how CGC changes over time. The authors describe the impact of genetic testing by targeted approach or universal approach on the management of BC according to the stage at diagnosis. Moreover, they describe the burden of CGC and testing and future perspectives to widely offer testing. A new perspective is needed for models of service delivery of CGC and testing, beyond formal genetic counselling. A broader genetic test can be quickly usable in clinical practice for comprehensive BC management and personalized prevention in the era of precision oncology.
有相当比例的乳腺癌(BC)与致病基因中的致病(P)/可能致病(LP)变异有关。对 P/LP 变异的了解是乳腺癌患者首次确诊后进行治疗的基本要素,因为它会影响手术和后续的肿瘤治疗及随访。此外,如果是 BRCA1/2 基因中 P/LP 变异的携带者,转移性 BC 患者可以从个性化治疗中获益。通过多基因检测,还可以发现其他对治疗有影响的易感基因。对健康的家庭成员进行连锁基因检测,可以制定个性化的预防策略。在此,我们回顾了癌症遗传咨询(CGC)的进展和挑战。我们将重点放在针对遗传性 BC 管理的肿瘤学领域,描述引导 CGC 的独特方式以及 CGC 如何随着时间的推移而变化。作者描述了根据诊断阶段,通过有针对性的方法或通用方法进行基因检测对 BC 管理的影响。此外,他们还描述了CGC和检测的负担以及广泛提供检测的未来前景。除了正规的遗传咨询外,还需要从新的视角看待提供遗传咨询和检测服务的模式。在精准肿瘤学时代,更广泛的基因检测可迅速用于临床实践,以实现对乳腺癌的全面管理和个性化预防。
{"title":"Cancer genetic counselling for hereditary breast cancer in the era of precision oncology","authors":"M. Pensabene , A. Calabrese , C. von Arx, R. Caputo, M. De Laurentiis","doi":"10.1016/j.ctrv.2024.102702","DOIUrl":"https://doi.org/10.1016/j.ctrv.2024.102702","url":null,"abstract":"<div><p>A relevant percentage of breast cancers (BCs) are tied to pathogenetic (P)/likely pathogenetic (LP) variants in predisposing genes. The knowledge of P/LP variants is an essential element in the management of BC patients since the first diagnosis because it influences surgery and subsequent oncological treatments and follow-up. Moreover, patients with metastatic BCs can benefit from personalized treatment if carriers of P/LP in BRCA1/2 genes. Multigene panels allow the identification of other predisposing genes with an impact on management. Cascade genetic testing for healthy family members allows personalized preventive strategies. Here, we review the advances and the challenges of Cancer Genetic Counseling (CGC). We focus on the area of oncology directed to hereditary BC management describing the peculiar way to lead CGC and how CGC changes over time. The authors describe the impact of genetic testing by targeted approach or universal approach on the management of BC according to the stage at diagnosis. Moreover, they describe the burden of CGC and testing and future perspectives to widely offer testing. A new perspective is needed for models of service delivery of CGC and testing, beyond formal genetic counselling. A broader genetic test can be quickly usable in clinical practice for comprehensive BC management and personalized prevention in the era of precision oncology.</p></div>","PeriodicalId":9537,"journal":{"name":"Cancer treatment reviews","volume":"125 ","pages":"Article 102702"},"PeriodicalIF":11.8,"publicationDate":"2024-02-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0305737224000203/pdfft?md5=df11a66a33e2c56e6989728f2a516d15&pid=1-s2.0-S0305737224000203-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140041819","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-02-27DOI: 10.1016/j.ctrv.2024.102701
Michele Bartoletti , Marcella Montico , Domenica Lorusso , Roberta Mazzeo , Ana Oaknin , Lucia Musacchio , Giovanni Scambia , Fabio Puglisi , Sandro Pignata
Importance
Various randomized trials have explored the efficacy of combining immune checkpoint inhibitors (ICIs) with first-line chemotherapy in advanced endometrial cancer. We aimed to summarize available data and clarify the benefit of adding immunotherapy according to the DNA mismatch repair status (deficient, dMMR or proficient, pMMR) and the specific type of agent used (anti-PD1 or anti-PD-L1).
Objective
To assess whether the addition of ICIs to standard platinum-based chemotherapy enhances progression-free survival (PFS) for patients with advanced endometrial cancer both overall and based on DNA mismatch repair status.
Data sources
Electronic databases (PubMed, Embase and Cochrane Library) and conference proceedings were searched for first line, randomized and controlled trials integrating ICIs with chemotherapy for the treatment of advanced endometrial cancer published or presented by November 1, 2023.
Study selection
Five studies, comprising 2456 patients (1308 received ICIs with chemotherapy and 1148 treated with chemotherapy alone) met the selection criteria and were included in the analysis. Experimental arms included pembrolizumab, dostarlimab (anti-PD1) and durvalumab, atezolizumab and avelumab (anti-PD-L1) combined with standard three-weekly carboplatin-paclitaxel chemotherapy backbone. Endometrial carcinosarcoma were included in 3 out of 5 trials.
Data extraction and synthesis
For comparison of PFS outcomes, extrapolation of hazard ratios (HRs), 95% confidence intervals (CI) and PFS events was performed for each included study in the overall population and according to subgroups. Data analysis was conducted using a random-effects model.
Results
The addition of ICIs to chemotherapy improved PFS compared to chemotherapy alone in the overall population (pooled HR, 0.63; 95 % CI, 0.52––0.76; P <.001). In the dMMR subgroup the benefit was more pronounced (pooled HR, 0.34; 95 % CI, 0.27––0.44; P <.001) and not affected by drugs used with pooled HRs of 0.39 (95 % CI, 0.28––0.55; P <.001) and 0.34 (95 % CI, 0.27––0.44; P <.001) for PD-L1 and PD1 inhibitors, respectively. For pMMR patients, a statistically significant benefit in terms of PFS was confirmed only when anti-PD1 were used (anti-PD-1: HR 0.64, 95 % CI: 0.46–0.90, P =.010 vs anti-PD-L1: HR 0.87, 95 % CI: 0.73–1.03, P =.104)
Conclusions and relevance
This meta-analysis confirmed the advantage in terms of PFS of adding ICIs to standard platinum-based chemotherapy. While dMMR patients benefit from the incorporation of both anti PD-1 or anti PD-L1, this benefit is confined to the association of anti-PD1 agents in pMMR patients. Updated analysis of trials is awaited to clarify the impact of immunotherapy on overall survival.
{"title":"Incorporation of anti-PD1 or anti PD-L1 agents to platinum-based chemotherapy for the primary treatment of advanced or recurrent endometrial cancer. A meta-analysis","authors":"Michele Bartoletti , Marcella Montico , Domenica Lorusso , Roberta Mazzeo , Ana Oaknin , Lucia Musacchio , Giovanni Scambia , Fabio Puglisi , Sandro Pignata","doi":"10.1016/j.ctrv.2024.102701","DOIUrl":"https://doi.org/10.1016/j.ctrv.2024.102701","url":null,"abstract":"<div><h3>Importance</h3><p>Various randomized trials have explored the efficacy of combining immune checkpoint inhibitors (ICIs) with first-line chemotherapy in advanced endometrial cancer. We aimed to summarize available data and clarify the benefit of adding immunotherapy according to the DNA mismatch repair status (deficient, dMMR or proficient, pMMR) and the specific type of agent used (anti-PD1 or anti-PD-L1).</p></div><div><h3>Objective</h3><p>To assess whether the addition of ICIs to standard platinum-based chemotherapy enhances progression-free survival (PFS) for patients with advanced endometrial cancer both overall and based on DNA mismatch repair status.</p></div><div><h3>Data sources</h3><p>Electronic databases (PubMed, Embase and Cochrane Library) and conference proceedings were searched for first line, randomized and controlled trials integrating ICIs with chemotherapy for the treatment of advanced endometrial cancer published or presented by November 1, 2023.</p></div><div><h3>Study selection</h3><p>Five studies, comprising 2456 patients (1308 received ICIs with chemotherapy and 1148 treated with chemotherapy alone) met the selection criteria and were included in the analysis. Experimental arms included pembrolizumab, dostarlimab (anti-PD1) and durvalumab, atezolizumab and avelumab (anti-PD-L1) combined with standard three-weekly carboplatin-paclitaxel chemotherapy backbone. Endometrial carcinosarcoma were included in 3 out of 5 trials.</p></div><div><h3>Data extraction and synthesis</h3><p>For comparison of PFS outcomes, extrapolation of hazard ratios (HRs), 95% confidence intervals (CI) and PFS events was performed for each included study in the overall population and according to subgroups. Data analysis was conducted using a random-effects model.</p></div><div><h3>Results</h3><p>The addition of ICIs to chemotherapy improved PFS compared to chemotherapy alone in the overall population (pooled HR, 0.63; 95 % CI, 0.52––0.76; P <.001). In the dMMR subgroup the benefit was more pronounced (pooled HR, 0.34; 95 % CI, 0.27––0.44; P <.001) and not affected by drugs used with pooled HRs of 0.39 (95 % CI, 0.28––0.55; P <.001) and 0.34 (95 % CI, 0.27––0.44; P <.001) for PD-L1 and PD1 inhibitors, respectively. For pMMR patients, a statistically significant benefit in terms of PFS was confirmed only when anti-PD1 were used (anti-PD-1: HR 0.64, 95 % CI: 0.46–0.90, P =.010 vs anti-PD-L1: HR 0.87, 95 % CI: 0.73–1.03, P =.104)</p></div><div><h3>Conclusions and relevance</h3><p>This <em>meta</em>-analysis confirmed the advantage in terms of PFS of adding ICIs to standard platinum-based chemotherapy. While dMMR patients benefit from the incorporation of both anti PD-1 or anti PD-L1, this benefit is confined to the association of anti-PD1 agents in pMMR patients. Updated analysis of trials is awaited to clarify the impact of immunotherapy on overall survival.</p></div>","PeriodicalId":9537,"journal":{"name":"Cancer treatment reviews","volume":"125 ","pages":"Article 102701"},"PeriodicalIF":11.8,"publicationDate":"2024-02-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0305737224000197/pdfft?md5=70aba726898887fdf324df961a3425eb&pid=1-s2.0-S0305737224000197-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139993345","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-02-24DOI: 10.1016/j.ctrv.2024.102700
David Johnson , Cheng Ean Chee , Wesley Wong , Rachel C.T. Lam , Iain Bee Huat Tan , Brigette B.Y. Ma
The last two decades have witnessed major breakthroughs in the development of targeted therapy for patients with metastatic colorectal cancer (mCRC), an achievement which stems largely from advances in translational research. Precision medicine is now widely practiced in routine oncological care, where systemic therapy is individualized based on clinical factors such as primary tumor sidedness, location and number of metastases, as well as molecular factors such as the RAS and BRAF mutation status, mismatch repair / microsatellite status and presence of other actionable genomic alterations in the tumor. The optimal selection of patients with RAS and BRAF-wild type (WT), left-sided primary tumor for treatment with epidermal growth factor receptor (EGFR) and chemotherapy (chemo) has markedly improved survival in the first-line setting. The pivotal trials of cetuximab in combination with BRAF/ MEK inhibitor for BRAF V600E mutant mCRC, and panitumumab with KRAS G12C inhibitor in KRAS(G12C)-mutant mCRC have been practice-changing. Anti-HER2 small molecular inhibitor, antibodies and antibody-drug conjugates have significantly improved the treatment outcome of patients with HER2 amplified mCRC. Anti-angiogenesis agents are now used across all lines of treatment and novel combinations with immune-checkpoint inhibitors are under active investigation in MSS mCRC. The non-invasive monitoring of molecular resistance to targeted therapies using Next Generation Sequencing analysis of circulating tumor-derived DNA (ctDNA) and captured sequencing of tumors have improved patient selection for targeted therapies. This review will focus on how latest advances, challenges and future directions in the development of targeted therapies in mCRC.
{"title":"Current advances in targeted therapy for metastatic colorectal cancer – Clinical translation and future directions","authors":"David Johnson , Cheng Ean Chee , Wesley Wong , Rachel C.T. Lam , Iain Bee Huat Tan , Brigette B.Y. Ma","doi":"10.1016/j.ctrv.2024.102700","DOIUrl":"https://doi.org/10.1016/j.ctrv.2024.102700","url":null,"abstract":"<div><p>The last two decades have witnessed major breakthroughs in the development of targeted therapy for patients with metastatic colorectal cancer (mCRC), an achievement which stems largely from advances in translational research. Precision medicine is now widely practiced in routine oncological care, where systemic therapy is individualized based on clinical factors such as primary tumor sidedness, location and number of metastases, as well as molecular factors such as the RAS and BRAF mutation status, mismatch repair / microsatellite status and presence of other actionable genomic alterations in the tumor. The optimal selection of patients with RAS and BRAF-wild type (WT), left-sided primary tumor for treatment with epidermal growth factor receptor (EGFR) and chemotherapy (chemo) has markedly improved survival in the first-line setting. The pivotal trials of cetuximab in combination with BRAF/ MEK inhibitor for BRAF V600E mutant mCRC, and panitumumab with KRAS G12C inhibitor in KRAS(G12C)-mutant mCRC have been practice-changing. Anti-HER2 small molecular inhibitor, antibodies and antibody-drug conjugates have significantly improved the treatment outcome of patients with HER2 amplified mCRC. Anti-angiogenesis agents are now used across all lines of treatment and novel combinations with immune-checkpoint inhibitors are under active investigation in MSS mCRC. The non-invasive monitoring of molecular resistance to targeted therapies using Next Generation Sequencing analysis of circulating tumor-derived DNA (ctDNA) and captured sequencing of tumors have improved patient selection for targeted therapies. This review will focus on how latest advances, challenges and future directions in the development of targeted therapies in mCRC.</p></div>","PeriodicalId":9537,"journal":{"name":"Cancer treatment reviews","volume":"125 ","pages":"Article 102700"},"PeriodicalIF":11.8,"publicationDate":"2024-02-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139993344","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-02-17DOI: 10.1016/j.ctrv.2024.102699
Emilio Francesco Giunta , Nicole Brighi , Giorgia Gurioli , Federica Matteucci , Giovanni Paganelli , Ugo De Giorgi
177Lu-PSMA has been approved for the treatment of PSMA-positive metastatic castration-resistant (mCRPC) patients who progressed to androgen receptor pathway inhibitors (ARPIs) and taxane-based chemotherapy. However, a higher proportion of patients do not respond to this type of radioligand therapy (RLT). To date, there is a lack of validated prognostic and predictive biomarkers for 177Lu-PSMA therapy in prostate cancer. Several studies have investigated the prognostic and predictive role of clinical and molecular factors and also the metabolic features of PET imaging. In this review, we aim to take stock of the current scenario, focusing on new emerging data from retrospective/prospective series and clinical trials. Given the high costs and the possibility of primary resistance, it seems essential to identify clinical and molecular characteristics that could allow clinicians to choose the right patient to treat with 177Lu-PSMA. Biomarker-based clinical trials are urgently needed in this field.
177Lu-PSMA已被批准用于治疗对雄激素受体途径抑制剂(ARPI)和以类固醇为基础的化疗有进展的PSMA阳性转移性阉割耐药(mCRPC)患者。然而,较高比例的患者对这种放射性配体疗法(RLT)没有反应。迄今为止,前列腺癌的 177Lu-PSMA 治疗缺乏有效的预后和预测生物标志物。有几项研究调查了临床和分子因素的预后和预测作用,以及 PET 成像的代谢特征。在这篇综述中,我们旨在总结目前的情况,重点关注来自回顾性/前瞻性系列研究和临床试验的新数据。考虑到高昂的费用和原发性耐药的可能性,确定临床和分子特征似乎至关重要,这可以让临床医生选择合适的患者进行 177Lu-PSMA 治疗。这一领域迫切需要基于生物标志物的临床试验。
{"title":"177Lu-PSMA therapy in metastatic prostate cancer: An updated review of prognostic and predictive biomarkers","authors":"Emilio Francesco Giunta , Nicole Brighi , Giorgia Gurioli , Federica Matteucci , Giovanni Paganelli , Ugo De Giorgi","doi":"10.1016/j.ctrv.2024.102699","DOIUrl":"10.1016/j.ctrv.2024.102699","url":null,"abstract":"<div><p>177Lu-PSMA has been approved for the treatment of PSMA-positive metastatic castration-resistant (mCRPC) patients who progressed to androgen receptor pathway inhibitors (ARPIs) and taxane-based chemotherapy. However, a higher proportion of patients do not respond to this type of radioligand therapy (RLT). To date, there is a lack of validated prognostic and predictive biomarkers for 177Lu-PSMA therapy in prostate cancer. Several studies have investigated the prognostic and predictive role of clinical and molecular factors and also the metabolic features of PET imaging. In this review, we aim to take stock of the current scenario, focusing on new emerging data from retrospective/prospective series and clinical trials. Given the high costs and the possibility of primary resistance, it seems essential to identify clinical and molecular characteristics that could allow clinicians to choose the right patient to treat with 177Lu-PSMA. Biomarker-based clinical trials are urgently needed in this field.</p></div>","PeriodicalId":9537,"journal":{"name":"Cancer treatment reviews","volume":"125 ","pages":"Article 102699"},"PeriodicalIF":11.8,"publicationDate":"2024-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0305737224000173/pdfft?md5=160030d37532638066675fd576081b44&pid=1-s2.0-S0305737224000173-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139927631","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}