Stephanie M Wang, Julia Dexter, Rebecca Wolsky, Carolyn Lefkowits
{"title":"Management of Advanced Stage Endometrial Cancer With Non-Measurable Disease After Surgery.","authors":"Stephanie M Wang, Julia Dexter, Rebecca Wolsky, Carolyn Lefkowits","doi":"10.1007/s11864-025-01345-1","DOIUrl":"10.1007/s11864-025-01345-1","url":null,"abstract":"<p><strong>Opinion statement: </strong>Patients with advanced stage completely resected endometrial cancer represent a heterogeneous group and decision-making regarding adjuvant treatment for this patient population is complex. When considering this cohort of patients, factors such as histologic subtype and molecular classification impact decision-making on adjuvant therapy. Options include systemic chemotherapy with or without immunotherapy or HER2-targeted therapy, chemoradiotherapy and radiation alone. We recommend tailoring treatment for patients with advanced-stage, non-measurable endometrial cancer based on HER2 status, mismatch repair (MMR) proficiency, disease stage and histology. For HER2-positive cases, we recommend carboplatin, paclitaxel, and trastuzumab. For HER2-negative, MMR-proficient disease, Stage 3 patients we recommend that have stage IIIB or IIIC disease AND grade 1 or 2 endometrioid histology OR grade 3 endometrioid histology p53 wild-type, we recommend carboplatin and paclitaxel, with consideration of the addition of pelvic radiationFor patients with stage IIIA disease OR grade 3 p53 aberrant endometrioid OR non-endometrioid histology, we recommend carboplatin and paclitaxel, with consideration of brachytherapy for patients with uterine risk factors such as lymphovascular space invasion, cervical stromal involvement, or lower uterine segment involvement. For Stage 4 patients with Her2 negative, MMRp disease, we recommend carboplatin and paclitaxel, with optional brachytherapy. For HER2-negative, MMR-deficient disease, the recommended regimen includes carboplatin, paclitaxel, and immunotherapy, with consideration of brachytherapy for patients with the uterine risk factors listed above.</p>","PeriodicalId":50600,"journal":{"name":"Current Treatment Options in Oncology","volume":" ","pages":"820-828"},"PeriodicalIF":4.7,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144976751","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Updates on Therapy Options in Fit and Unfit Patients with Newly Diagnosed AML.","authors":"Gray H Magee, Michael R Grunwald","doi":"10.1007/s11864-025-01351-3","DOIUrl":"10.1007/s11864-025-01351-3","url":null,"abstract":"<p><strong>Opinion statement: </strong>The integration of next-generation sequencing (NGS) and advanced cytogenetic diagnostics into routine clinical practice is reshaping frontline treatment of acute myeloid leukemia (AML) in both fit and unfit patients. Molecular profiling now enables personalized treatment strategies, particularly for patients harboring mutations in FLT3, IDH1, IDH2, KMT2A, and NPM1. Small molecule inhibitors, first reserved for relapsed/refractory disease, are increasingly used in the upfront setting. However, universal NGS testing at diagnosis is critical to identify eligible patients for these targeted therapies. In patients lacking actionable mutations, treatment can still be refined using karyotypic abnormalities or high-risk features suggestive of antecedent MDS. In our practice, we continue to use 7 + 3 induction for fit patients, adding midostaurin or quizartinib for FLT3-mutated AML, or gemtuzumab ozogamicin for core binding factor (CBF) AML expressing CD33. For patients with therapy-related AML or AML with myelodysplasia-related changes, CPX-351 is our standard induction approach. For unfit patients, we generally offer hypomethylating agents with venetoclax. In the presence of IDH1 mutations, we consider azacitidine combined with ivosidenib. If venetoclax is contraindicated or not tolerated, targeted therapies such as gilteritinib, ivosidenib, or enasidenib may be appropriate based on mutation profile. However, we try to identify clinical trials for all our patients at diagnosis. One of the more exciting recent developments is the emergence of menin inhibitors for patients with KMT2A rearrangements or NPM1 mutations. While several agents have received FDA approval or breakthrough status in the relapsed/refractory setting, they are now being actively studied as frontline options with promising results. When feasible, clinical trial enrollment should be considered for newly diagnosed patients with these alterations. As the therapeutic landscape for AML continues to evolve, timely molecular characterization is more essential than ever to optimize outcomes and select the most appropriate frontline strategy.</p>","PeriodicalId":50600,"journal":{"name":"Current Treatment Options in Oncology","volume":" ","pages":"829-840"},"PeriodicalIF":4.7,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12449387/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144976802","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Navigating PARP Inhibitor Resistance in Ovarian Cancer: Bridging Mechanistic Insights To Clinical Translation.","authors":"Ziyi Wang, Yuting Liu, Qing Yang","doi":"10.1007/s11864-025-01347-z","DOIUrl":"10.1007/s11864-025-01347-z","url":null,"abstract":"<p><strong>Opinion statement: </strong>PARP inhibitors (PARPi) are increasingly vital in treating, particularly in individuals exhibiting homologous recombination deficiency (HRD), harnessing synthetic lethality to target tumor cells. However, PARPi resistance severely restricts their sustained efficacy, posing a significant clinical obstacle requiring a detailed exploration of its mechanisms and solutions. This review elucidates the underlying mechanisms of PARPi resistance, assesses biomarker-driven predictive tools, and explores novel strategies to improve therapeutic outcomes. This review critically assesses biomarker-driven strategies for resistance prediction, such as genomic profiling, functional assays, dynamic circulating tumor DNA (ctDNA) monitoring, and emerging markers, to refine patient stratification. Moreover, this review systematically dissects PARPi resistance, including genomic alterations, homologous recombination (HR) restoration, replication fork stabilization, enhanced drug efflux, reduced PARP1 trapping, and microenvironmental influences. In addition, therapeutic tactics to prevent or overcome resistance are evaluated, encompassing sequential PARPi use, combinations with chemotherapy, immunotherapy, antiangiogenic agents, and DNA damage response (DDR) inhibitors, alongside innovative approaches like antibody-drug conjugates (ADCs). Bridging mechanistic and clinical perspectives, this review promotes a multidisciplinary approach to optimize PARPi-based treatments, addressing current challenges in ovarian cancer management.</p>","PeriodicalId":50600,"journal":{"name":"Current Treatment Options in Oncology","volume":" ","pages":"797-819"},"PeriodicalIF":4.7,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144876630","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Emerging Therapeutic Approaches to Pancreatic Adenocarcinoma: Advances and Future Directions.","authors":"Chengwei Peng, Paul E Oberstein","doi":"10.1007/s11864-025-01352-2","DOIUrl":"https://doi.org/10.1007/s11864-025-01352-2","url":null,"abstract":"<p><strong>Opinion statement: </strong>Pancreatic adenocarcinoma remains a leading cause of cancer-related mortality worldwide. Although surgery can be curable for a subset of patients, the five-year overall survival remains less than 15%. Despite extensive molecular characterization of pancreatic cancer, cytotoxic chemotherapy has served as the major component in therapeutic management. A major driver of pancreatic adenocarcinoma is mutations in KRAS, present in over 90% of cases. However, attempts to inhibit KRAS through upstream and downstream targets through the mitogen-activated protein kinase pathway have not been successful in the past. Despite this, multiple KRAS inhibitors have recently entered clinical trials and have shown promising results. These inhibitors have the potential to dramatically alter the landscape of treatment. In parallel, immunological approaches utilizing vaccines and bispecific antibodies are also in clinical development. Given these rapid new developments, the future of pancreatic cancer treatment will likely be determined by discovering the appropriate combinations of targeted and immune-based treatments.</p>","PeriodicalId":50600,"journal":{"name":"Current Treatment Options in Oncology","volume":" ","pages":""},"PeriodicalIF":4.7,"publicationDate":"2025-08-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144976570","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Covalent and Non-Covalent BTK Inhibition in Chronic Lymphocytic Leukemia Treatment.","authors":"Pratik V Shah, Douglas E Gladstone","doi":"10.1007/s11864-025-01339-z","DOIUrl":"10.1007/s11864-025-01339-z","url":null,"abstract":"<p><strong>Opinion statement: </strong>Bruton's tyrosine kinase (BTK) inhibitors are transforming chronic lymphocytic leukemia (CLL) treatment. Given the availability of both covalent and non-covalent BTK inhibitors (BTKis) CLL treatment has become more nuanced. Here the mechanisms of action, efficacy, and resistance patterns associated with both inhibitor classes are reviewed to help create a framework for integrating covalent and non-covalent BTKis into CLL treatment algorithms. Our treatment algorithm based on the available data is as follows, in the frontline setting when oral therapy is preferred/chosen and indefinite therapy is not a deterrent, irrespective of DNA and IGVH mutational status, a second-generation covalent BTK inhibitor (cBTKi) is recommended. In the relapsed/refractory setting when oral therapy is preferred a second-generation cBTKi is preferred. For those suffering disease progression during second generation cBTK, it is recommended to change to an alternative cBTKi and for those exposed to greater than two lines of therapy, challenging to a non-covalent BTKi (ncBTKi) in the form of pirtobrutinib. At the time of disease progression on a BTKi, we recommend testing for resistance mutations. Data on combination therapy's role and time-limited BTKi use remain under active investigation.</p>","PeriodicalId":50600,"journal":{"name":"Current Treatment Options in Oncology","volume":" ","pages":"754-763"},"PeriodicalIF":4.7,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144660965","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Imperia Nuzzolese, Stefano Cavalieri, Lisa F Licitra, Salvatore Alfieri
{"title":"The Role of Cetuximab in Non-Melanoma Skin Cancer: A Review of Clinical Evidence and Emerging Strategies.","authors":"Imperia Nuzzolese, Stefano Cavalieri, Lisa F Licitra, Salvatore Alfieri","doi":"10.1007/s11864-025-01335-3","DOIUrl":"10.1007/s11864-025-01335-3","url":null,"abstract":"<p><p>Non-melanoma skin cancers (NMSCs), particularly basal cell carcinoma (BCC) and cutaneous squamous cell carcinoma (cSCC), are the most common malignancies in the Caucasian population. While localized disease has a high cure rate, advanced cases pose therapeutic challenges. The introduction of immune checkpoint inhibitors (ICIs) has revolutionized treatment; however, resistance and contraindications necessitate alternative strategies. Epidermal growth factor receptor (EGFR) inhibition, particularly with cetuximab, has emerged as a potential therapeutic option in cSCC. This review evaluates the role of cetuximab in NMSC, focusing on clinical efficacy, safety, and emerging therapeutic strategies. A literature search was conducted using PubMed and clinical trial databases to identify relevant studies on cetuximab in advanced cSCC and BCC. The review discusses its use as monotherapy, in combination with radiotherapy or chemotherapy, and in the post-ICI setting. Despite the dominance of ICIs in advanced cSCC, cetuximab remains a valuable option, particularly for patients with ICI failure or contraindications. Its role as a radiosensitizer and its potential combination with immunotherapy warrant further investigation. Future studies should clarify optimal sequencing and combination strategies to improve patient outcomes.</p>","PeriodicalId":50600,"journal":{"name":"Current Treatment Options in Oncology","volume":" ","pages":"673-687"},"PeriodicalIF":4.7,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144592856","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Cancer-Related Alopecia Risk and Treatment.","authors":"Lily Kaufman, Lilia Valentic, Hannah Moulton, Lucy Rose, Brittany Dulmage","doi":"10.1007/s11864-025-01336-2","DOIUrl":"10.1007/s11864-025-01336-2","url":null,"abstract":"<p><strong>Opinion statement: </strong>Cancer-related alopecia (CRA) presents a significant challenge for many patients undergoing cancer treatment, often affecting their psychological well-being and sense of identity. In my opinion, the optimal management of CRA requires a proactive, personalized approach that prioritizes both prevention and regrowth, while taking into account the type of cancer therapy, patient goals, and overall clinical context. For patients receiving chemotherapy, especially taxane- or anthracycline-based regimens, scalp cooling should be offered as a first-line preventative option whenever feasible. Its demonstrated effectiveness, particularly when appropriately sequenced with chemotherapy agents, makes it a valuable tool in preserving hair and quality of life. For patients with contraindications to scalp cooling or limited access to this intervention, early counseling and support around hair loss expectations and coping strategies remain critical. In terms of regrowth, topical minoxidil remains the most evidence-based pharmacologic option and should be recommended, especially for patients with endocrine therapy- or chemotherapy-induced alopecia. While oral minoxidil shows promise, it should be used with caution until more robust safety data are available in oncology settings. Spironolactone, tretinoin, prostaglandin analogs, and red light therapy may be considered in select cases, especially when standard options are insufficient, though patients should be counseled on the limitations of available evidence. Ultimately, a patient-centered, multidisciplinary approach is key to optimizing outcomes in CRA care.</p>","PeriodicalId":50600,"journal":{"name":"Current Treatment Options in Oncology","volume":" ","pages":"706-715"},"PeriodicalIF":4.7,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12408754/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144627640","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"A Milestone in the Shift from \"Passive Killing\" to \"Active Immunomodulation\" in Cancer Treatment-Progress in Melanoma Vaccine Research.","authors":"Yuke Zhang, Jie Liu, Xiyue Chang, Xuejing Yang, Xinyue Zhang, Wan'an Xiao","doi":"10.1007/s11864-025-01340-6","DOIUrl":"10.1007/s11864-025-01340-6","url":null,"abstract":"<p><strong>Opinion statement: </strong>Melanoma is the most malignant skin tumor and exhibits extremely aggressive behavior with a high tendency to metastasize. Although the 5-year survival rate of early-stage patients (stage I-II) can reach over 90%, intermediate- and late-stage patients (stage III-IV) still face a high risk of recurrence and metastasis even after surgical resection. Moreover, these patients show low sensitivity to traditional therapies such as chemotherapy and radiotherapy. While immune checkpoint therapies (ICIs) represented by PD-1 inhibitors have significantly improved patient prognosis, they remain ineffective in 30%-40% of cases due to tumor immune escape or microenvironmental suppression. Additionally, long-term use of ICIs may induce autoimmune side effects (e.g., thyroid dysfunction). Therefore, there is an urgent need to develop more precise and durable treatment strategies. Vaccines for the treatment of melanoma represent one of the most promising therapeutic approaches. Melanoma vaccines activate the immune system through antigen recognition, leading to precise tumor killing. The vaccine also induces immune memory, demonstrates safety, and can be combined with immune checkpoint inhibitors to significantly enhance efficacy-making it a vital treatment for postoperative high-risk patients in their pursuit of long-term disease-free survival. With theoretical breakthroughs and technological iterations in immunotherapy, the development of melanoma vaccines has thrived and shown encouraging efficacy in clinical trials. Current mainstream melanoma vaccines are categorized into cell-based, peptide, nucleic acid, and viral types. Remaining challenges include complex and costly production processes, individual variability in therapeutic response, limited efficacy in advanced-stage patients, and potential autoimmune risks. Although melanoma vaccines face many shortcomings and challenges, these difficulties are being actively addressed through innovative strategies. For example, mRNA vaccine optimization involves improving stability and immunogenicity while shortening preparation cycles via enhanced lipid nanoparticle delivery and antigen design; novel vaccine platforms include nanovaccines and phage vaccines; combination therapy strategies entail combining vaccines with immune checkpoint inhibitors or targeted therapies; and using multi-omics analysis and AI to predict neoantigens, screen patients most likely to benefit, and develop efficacy monitoring markers to optimize treatment regimens. Although most melanoma vaccines are still under research, their emergence exemplifies the realization of individualized precision medicine and marks a milestone in the shift from \"passive killing\" to \"active immunomodulation\" in cancer treatment. This study provides an overview of recent research on melanoma vaccines to enable readers to grasp the latest advances in this field.</p>","PeriodicalId":50600,"journal":{"name":"Current Treatment Options in Oncology","volume":" ","pages":"688-705"},"PeriodicalIF":4.7,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144660964","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hue T T Tran, Thuy M Tran, Duc V Le, Jena C Jacobs, Trang M Nguyen, Huy L Trinh, Binh T T Vo, Tung S Tran, Giang H Nguyen
{"title":"Skin Reactions and Other Underappreciated Dermatologic Side Effects of Cancer Therapies.","authors":"Hue T T Tran, Thuy M Tran, Duc V Le, Jena C Jacobs, Trang M Nguyen, Huy L Trinh, Binh T T Vo, Tung S Tran, Giang H Nguyen","doi":"10.1007/s11864-025-01333-5","DOIUrl":"10.1007/s11864-025-01333-5","url":null,"abstract":"<p><strong>Opinion statement: </strong>Cutaneous adverse events (cAEs) are among the most common toxicities associated with modern cancer therapies, which are particularly heightened among immunotherapies and targeted agents. Despite being frequently immune-mediated, the majority of cAEs are mild to moderate and can be effectively managed without interruption of anticancer treatment. In immunotherapies-receiving patients, common phenotypes of cAEs include eczema-like, lichenoid, psoriasiform, vitiligo-like, and bullous eruptions. Targeted therapies including epidermal growth factor receptor inhibitors, BRAF inhibitors/MEK inhibitors, and phosphoinositide 3-kinase inhibitors are frequently associated with papulopustular eruptions, xerosis, paronychia, and photosensitivity. Mechanistically, cAEs may result from on-target immune activation, which correlates with treatment efficacy, or off-target hypersensitivity. Notably, certain phenotypes such as vitiligo, alopecia areata, and lichenoid reactions have been associated with improved survival in melanoma and non-small cell lung cancer. Management is guided based on the Common Terminology Criteria for Adverse Events grading, emphasizing the role of topical therapies for mild cases, systemic corticosteroids or immunosuppressants for moderate-to-severe reactions, and biologic or novel topical agents for steroid-refractory disease. By timely and appropriate intervention, most cAEs are manageable and may carry favorable prognostic significance. Early dermatologic collaboration is essential to reduce morbidity and ensure uninterrupted oncologic therapy.</p>","PeriodicalId":50600,"journal":{"name":"Current Treatment Options in Oncology","volume":" ","pages":"726-753"},"PeriodicalIF":4.7,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144856951","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Re-thinking the Role of Thoracic Radiotherapy in the First-Line Treatment for Extensive-Stage Small Cell Lung Cancer.","authors":"Guo Lin, Fan Ge, Chao Yang, Ying Huang","doi":"10.1007/s11864-025-01342-4","DOIUrl":"10.1007/s11864-025-01342-4","url":null,"abstract":"<p><strong>Opinion statement: </strong>Extensive-stage small cell lung cancer (ES-SCLC) remains a challenging disease with a poor prognosis, despite recent advances in immunotherapy. The integration of thoracic radiotherapy (TRT) with chemoimmunotherapy has emerged as a promising strategy to improve treatment outcomes. However, the optimal role and timing of TRT in the first-line treatment of ES-SCLC in the immunotherapy era are still under investigation. Traditional approaches to TRT, based on high-dose regimens, have shown limited efficacy in overcoming the immunosuppressive tumor microenvironment (TME) characteristic of SCLC. The advent of low-dose radiotherapy (LDRT) offers a novel perspective by modulating the TME to enhance antitumor immunity. This review aims to re-evaluate the role of TRT, particularly LDRT, in the context of evolving immunotherapy strategies for ES-SCLC. By examining recent clinical trials and preclinical studies, we discuss the potential mechanisms through which LDRT can reshape the TME, promote immune activation, and improve the efficacy of immunotherapy. Additionally, we highlight the ongoing clinical trials that are essential for validating these approaches and determining the optimal treatment paradigms.</p>","PeriodicalId":50600,"journal":{"name":"Current Treatment Options in Oncology","volume":" ","pages":"764-774"},"PeriodicalIF":4.7,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144762216","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}