1
|
Pera MF. A brief chronicle of research on human pluripotent stem cells. Bioessays 2024:e2400092. [PMID: 39058898 DOI: 10.1002/bies.202400092] [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: 04/13/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024]
Abstract
Today, human pluripotent stem cell technologies find widespread application across biomedical research, as models for early human development, as platforms for functional human genomics, as tools for the study of disease, drug screening and toxicology, and as a renewable source of cellular therapeutics for a range of intractable diseases. The foundations of this human pluripotent stem cell revolution rest on advances in a wide range of disciplines, including cancer biology, assisted reproduction, cell culture and organoid technology, somatic cell nuclear transfer, primate embryology, single-cell biology, and gene editing. This review surveys the slow emergence of the study of human pluripotency and the exponential growth of the field during the past several decades.
Collapse
Affiliation(s)
- Martin F Pera
- JAX Mammalian Genetics, The Jackson Laboratory, Bar Harbor, Maine, USA
| |
Collapse
|
2
|
Sriram S, Macedo T, Mavinkurve‐Groothuis A, van de Wetering M, Looijenga LHJ. Alkylating agents-induced gonadotoxicity in prepubertal males: Insights on the clinical and preclinical front. Clin Transl Sci 2024; 17:e13866. [PMID: 38965809 PMCID: PMC11224131 DOI: 10.1111/cts.13866] [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: 04/12/2024] [Revised: 05/26/2024] [Accepted: 06/06/2024] [Indexed: 07/06/2024] Open
Abstract
Rising cure rates in pediatric cancer patients warrants an increased attention toward the long-term consequences of the diagnosis and treatment in survivors. Chemotherapeutic agents can be gonadotoxic, rendering them at risk for infertility post-survival. While semen cryopreservation is an option that can be provided for most (post)pubertal boys before treatment, this is unfortunately not an option prepubertal in age, simply due to the lack of spermatogenesis. Over the last couple of years, studies have thus focused on better understanding the testis niche in response to various chemotherapeutic agents that are commonly administered and their direct and indirect impact on the germ cell populations. These are generally compounds that have a high risk of infertility and have been classified into risk categories in curated fertility guidelines. However, with it comes the lack of evidence and the challenge of using informative models and conditions most reflective of the physiological scenario, in short, the appropriate study designs for clinically relevant outcomes. Besides, the exact mechanism(s) of action for many of these "risk" compounds as well as other agents is unclear. Understanding their behavior and effect on the testis niche will pave the way for incorporating new strategies to ultimately combat infertility. Of the various drug classes, alkylating agents pose the highest risk of gonadotoxicity as per previously established studies as well as risk stratification guidelines. Therefore, this review will summarize the findings in the field of male fertility concerning gonadotoxicity of akylating agents as a result of chemotherapy exposure.
Collapse
Affiliation(s)
- Sruthi Sriram
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Tiago Macedo
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | | | | |
Collapse
|
3
|
Hammami MB, Gudino P, Rodriguez Salazar JD, Vegivinti C, Qasim A, Acharya A. Assessing the prevalence and severity of cisplatin-induced nephrotoxicity in a minority- low socioeconomic population in the Bronx, New York. J Chemother 2024:1-6. [PMID: 38946180 DOI: 10.1080/1120009x.2024.2363105] [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: 01/25/2024] [Accepted: 05/30/2024] [Indexed: 07/02/2024]
Abstract
Studies evaluating Cisplatin-induced nephrotoxicity in minorities are limited. We conducted a retrospective review of adult patients receiving cisplatin from 2019 to 2023 at an inner-city hospital. Renal indices were obtained at baseline and after cycles 1, 2, and 3 of Cisplatin. A total of 93 patients were included, 46% were male. Median age was 57 years. About 40% were Black, 13% White, and 42% Hispanic. About 54% were uninsured. About 16% of the patients developed AKI after cycle 1 of cisplatin, 5% after cycle 2%, and 17% after cycle 3. There was no statistically significant correlation between race, sex, BMI and development of cisplatin-induced AKI. Repeated measures ANOVA test indicated a statistically significant and cumulative rise in creatinine level following cisplatin therapy [Wilks' Lambda = 0.003, F(1,26)=13.7, η2 = 0.44]. Our study in a minority, low socioeconomic population highlights the progressive kidney injury following each cycle of cisplatin therapy. Further studies targeting this specific population are warranted to develop tailored interventions.
Collapse
Affiliation(s)
- M Bakri Hammami
- Department of Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Paola Gudino
- Department of Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Charan Vegivinti
- Department of Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Asma Qasim
- Department of Medicine, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - Anjali Acharya
- Department of Nephrology, Jacobi Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
4
|
Jackson JC, Sanchez D, Johns AC, Campbell MT, Aydin AM, Gokden N, Maraboyina S, Muesse JL, Ward JF, Pisters LL, Zacharias NM, Guo CC, Tu SM. Germ Cell Tumor of the Testis: Lethal Subtypes of a Curable Cancer. J Clin Med 2024; 13:3436. [PMID: 38929965 PMCID: PMC11205088 DOI: 10.3390/jcm13123436] [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: 05/13/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Germ cell tumor of the testis (GCT) is a curable cancer even when it is widely metastatic; however, outcomes can differ based on tumor histology. Chemo-resistance in certain phenotypes, such as teratoma and yolk sac tumor, contributes to poor clinical outcomes in some patients with GCT. Despite this resistance to S-YSTemic therapy, many of these tumor subtypes remain amenable to surgical resection and possible cure. In this study, we report on a series of seven patients highlighting two chemo-resistant subtypes of nonseminomatous germ cell tumor (NSGCT), sarcomatoid yolk sac tumor (S-YST), and epithelioid trophoblastic tumor (ETT) for which early resection rather than additional salvage chemotherapy or high-dose intense chemotherapy might provide a superior clinical outcome and enhance cure rate.
Collapse
Affiliation(s)
- Jamaal C. Jackson
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.C.J.); (D.S.); (J.F.W.); (L.L.P.); (N.M.Z.)
| | - Darren Sanchez
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.C.J.); (D.S.); (J.F.W.); (L.L.P.); (N.M.Z.)
| | - Andrew C. Johns
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.C.J.); (M.T.C.)
| | - Matthew T. Campbell
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.C.J.); (M.T.C.)
| | - Ahmet M. Aydin
- Division of Urology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Neriman Gokden
- Division of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Sanjay Maraboyina
- Division of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jason L. Muesse
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - John F. Ward
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.C.J.); (D.S.); (J.F.W.); (L.L.P.); (N.M.Z.)
| | - Louis L. Pisters
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.C.J.); (D.S.); (J.F.W.); (L.L.P.); (N.M.Z.)
| | - Niki M. Zacharias
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.C.J.); (D.S.); (J.F.W.); (L.L.P.); (N.M.Z.)
| | - Charles C. Guo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Shi-Ming Tu
- Division of Hematology/Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
5
|
Ebel L, Autrán AM, Rodríguez-Faba O, Alcántara C. Testicular cancer in Ibero-America: Proper prevention and diagnostic strategies. Actas Urol Esp 2024:S2173-5786(24)00079-9. [PMID: 38848949 DOI: 10.1016/j.acuroe.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024]
Affiliation(s)
- L Ebel
- Escuela de Medicina Valdivia, Chile.
| | - A M Autrán
- Universidad Veracruzana, Veracrux, Mexico
| | | | - C Alcántara
- Hospital de Amor, Dr. Paulo Prata, Barretos, Brazil
| |
Collapse
|
6
|
Xu Y, Xie S, Zhou C, Zhu L, Tong Y, Munoz A, Wu Y, Li X. Time trends in the mortality of testicular cancer across the BRICS: an age-period-cohort analysis for the GBD 2019. Sci Rep 2024; 14:12740. [PMID: 38830945 PMCID: PMC11148183 DOI: 10.1038/s41598-024-63191-9] [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: 12/19/2023] [Accepted: 05/27/2024] [Indexed: 06/05/2024] Open
Abstract
Testicular cancer (TCa) is a rare but impactful malignancy that primarily affects young men. Understanding the mortality rate of TCa is crucial for improving prevention and treatment strategies to reduce the risk of death among patients. We obtained TCa mortality data by place (5 countries), age (20-79 years), and year (1990-2019) from the Global Burden of Disease Study 2019. Age-period-cohort model was used to estimate the net drift, local drift, age effects, period and cohort effects. In 2019, the global mortality of TCa increased to 10842 (95% UI 9961, 11902), with an increase of 50.08% compared to 1990.The all-age mortality rate for TCa in 2019 increased from 0.17/100,000 (95% UI 0.13, 0.20) in China to 0.48/100,000 (95% UI 0.38, 0.59) in Russian Federation, whereas the age-standardized mortality rate in 2019 was highest in the South Africa 0.47/100,000 (95% UI 0.42, 0.53) and lowest in the China 0.16/100,000 (95% UI 0.13, 0.19). China's aging population shifts mortality patterns towards the elderly, while in Russian Federation, young individuals are primarily affected by the distribution of deaths. To address divergent TCa mortality advancements in BRICS countries, we propose a contextually adaptive and resource-conscious approach to prioritize TCa prevention. Tailoring strategies to contextual diversity, including policy frameworks, human resources, and financial capacities, will enhance targeted interventions and effectiveness in reducing TCa mortality.
Collapse
Affiliation(s)
- Yuting Xu
- Department of Rehabilitation Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shudong Xie
- Transplantation Center, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chengyu Zhou
- Department of Rehabilitation Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liping Zhu
- Department of Rehabilitation Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yao Tong
- Department of Rehabilitation Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Alvaro Munoz
- Centro Universitario del Norte, Universidad de Guadalajara, Colotlán, Jalisco, Mexico
| | - Yuhang Wu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China.
| | - Xuhong Li
- Department of Rehabilitation Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
7
|
Wessman S, Nistér M, Kokaraki G, Pal N, Tettamanti G, Petta TB, Carlson JW. A comprehensive population-based study of malignant ovarian tumors, including histologic and immunohistochemical review, in children and adolescents 0-19 years old in Sweden between 1970 and 2014. Gynecol Oncol 2024; 184:206-213. [PMID: 38340646 DOI: 10.1016/j.ygyno.2024.01.035] [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: 04/24/2023] [Revised: 07/21/2023] [Accepted: 01/22/2024] [Indexed: 02/12/2024]
Abstract
OBJECTIVES Ovarian tumors in the pediatric population are rare. The incidence and frequency of subtypes differ between children and adults. Although not all tumors are aggressive, they may still lead to morbidity. The goal of this study was a comprehensive review of malignant ovarian tumors in children and adolescents diagnosed and registered in Sweden. METHODS Individuals were identified through a search in the National Cancer Register, limited for ages 0-19, years 1970-2014. Stored tumor diagnostic material from regional biobanks was retrieved and reviewed. RESULTS The study includes 345 individuals with ovarian tumors and 70.7% of them were between 15 and 19 years at time of diagnosis. No differences in incidence over time or geographic location were identified. The average follow-up time was 21.2 years and 5-year survival was 88.4%. Survival was similar in the different time periods, except for 1970-1979. Review was possible for 260 cases, resulting in 85 epithelial tumors, 121 GCTs, 47 SCSTs and 7 others. For age 0-4 years SCSTs dominated (85.7%), for 5-9- and 10-14-years GCTs dominated (70,8% and 75.0% respectively), and for age 15-19 years epithelial tumors dominated (43.8%). There was a strong agreement between review diagnosis and original diagnosis (Cohen's κ 0.944). Differentiating between entities within the sex cord-stromal group posed the biggest diagnostic challenge. CONCLUSIONS Ovarian tumors in children and adolescents are rare and distinct from their adult counterparts regarding incidence and frequency. There was a strong concurrence between original and review diagnoses. The greatest diagnostic difficulty was subtyping of epithelial tumors and differentiating between tumors within the SCST group.
Collapse
Affiliation(s)
- Sandra Wessman
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Pathology and Cancer diagnostics, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Monica Nistér
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Pathology and Cancer diagnostics, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Georgia Kokaraki
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, 90033 Los Angeles, CA, USA
| | - Niklas Pal
- Department of Women's and Children's Health, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Pediatric Oncology, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Giorgio Tettamanti
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Institute of Environmental Medicine, Unit of Epidemiology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Tirzah Braz Petta
- Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, 90033 Los Angeles, CA, USA; Department of Cellular Biology and Genetics, Federal University of Rio Grande de Norte, Natal, RN 59078-970, Brazil
| | - Joseph W Carlson
- Department of Oncology-Pathology, Karolinska Institutet, SE-171 77 Stockholm, Sweden; Department of Pathology and Laboratory Medicine, Keck School of Medicine, University of Southern California, 90033 Los Angeles, CA, USA.
| |
Collapse
|
8
|
Saliyeva S, Boranbayeva R, Bulegenova M, Zhumadullayev B, Nurzhanova G, Manzhuova L. Outcomes of pediatric extracranial germ cell tumors: A single center experience in a developing country. Rare Tumors 2024; 16:20363613231216567. [PMID: 38566825 PMCID: PMC10986168 DOI: 10.1177/20363613231216567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 11/08/2023] [Indexed: 04/04/2024] Open
Abstract
The purpose of this study was to analyze the outcomes of extracranial GCT in children in a developing country and to assess prognostic factors. The data on 141 children (<18 years old) with extracranial GCT, confirmed histopathologically, collected over the past 9 years (from February 2013 to June 2022) were retrospectively studied. The patients underwent the same therapy with platinum-containing chemotherapy regimens. In the malignant GCT group, OS and EFS were 81.0 ± 4% and 73 ± 5%, respectively. OS and EFS in the teratoma group were 90 ± 5% and 85 ± 6%. In univariate analysis, parameters like stage of disease, tumor localization, AFP level ≥10,000 ng/mL, serum AFP kinetics and resection status were found to be statistically significant prognostic factors. In the multivariate analysis, the significant adverse factors were the resection status, initial AFP level ≥10,000 ng/mL and serum AFP kinetics slow down (p = .000). Good survival rates can be achieved in developing countries with adequate compliance with treatment protocols. The analysis demonstrates high efficacy of platinum-containing chemotherapy regimens. In our opinion, the protocol used in high-income countries can be implemented in low-income countries with the financial support from the government. The qualification of specialists is also important.
Collapse
Affiliation(s)
- Symbat Saliyeva
- Oncology/hematology Department, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
- Department of Children's Diseases, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Riza Boranbayeva
- Oncology/hematology Department, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
- Department of Children's Diseases, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Minira Bulegenova
- Oncology/hematology Department, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
- Department of Children's Diseases, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Bakhram Zhumadullayev
- Oncology/hematology Department, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
| | - Gaukhar Nurzhanova
- Oncology/hematology Department, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
| | - Lyazat Manzhuova
- Oncology/hematology Department, Scientific Center of Pediatrics and Pediatric Surgery, Almaty, Kazakhstan
- Department of Children's Diseases, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| |
Collapse
|
9
|
McHugh DJ, Gleeson JP, Feldman DR. Testicular cancer in 2023: Current status and recent progress. CA Cancer J Clin 2024; 74:167-186. [PMID: 37947355 DOI: 10.3322/caac.21819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/25/2023] [Accepted: 09/29/2023] [Indexed: 11/12/2023] Open
Abstract
Testicular germ cell tumor (GCT) is the most common solid tumor in adolescent and young adult men. Progress in the management of GCT has been made in the last 50 years, with a substantial improvement in cure rates for advanced disease, from 25% in the 1970s to nearly 80%. However, relapsed or platinum-refractory disease occurs in a proportion, 20% of whom will die from disease progression. This article reviews the current evidence-based treatments for extracranial GCT, the acute and chronic toxic effects that may result, and highlights contemporary advances and progress in the field.
Collapse
Affiliation(s)
- Deaglan J McHugh
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medicine, New York, New York, USA
| | - Jack P Gleeson
- Cancer Research, College of Medicine and Health, University College Cork, Cork, Ireland
- Medical Oncology Department, Cork University Hospital, Cork, Ireland
| | - Darren R Feldman
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
10
|
Bork T, Hernando-Erhard C, Liang W, Tian Z, Yamahara K, Huber TB. Cisplatin Nephrotoxicity Is Critically Mediated by the Availability of BECLIN1. Int J Mol Sci 2024; 25:2560. [PMID: 38473806 DOI: 10.3390/ijms25052560] [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: 01/18/2024] [Revised: 02/10/2024] [Accepted: 02/13/2024] [Indexed: 03/14/2024] Open
Abstract
Cisplatin nephrotoxicity is a critical limitation of solid cancer treatment. Until now, the complex interplay of various pathophysiological mechanisms leading to proximal tubular cell apoptosis after cisplatin exposure has not been fully understood. In our study, we assessed the role of the autophagy-related protein BECLIN1 (ATG6) in cisplatin-induced acute renal injury (AKI)-a candidate protein involved in autophagy and with putative impact on apoptosis by harboring a B-cell lymphoma 2 (BCL2) interaction site of unknown significance. By using mice with heterozygous deletion of Becn1, we demonstrate that reduced intracellular content of BECLIN1 does not impact renal function or autophagy within 12 months. However, these mice were significantly sensitized towards cisplatin-induced AKI, and by using Becn1+/-;Sglt2-Cre;Tomato/EGFP mice with subsequent primary cell analysis, we confirmed that nephrotoxicity depends on proximal tubular BECLIN1 content. Mechanistically, BECLIN1 did not impact autophagy or primarily the apoptotic pathway. In fact, a lack of BECLIN1 sensitized mice towards cisplatin-induced ER stress. Accordingly, the ER stress inhibitor tauroursodeoxycholic acid (TUDCA) blunted cisplatin-induced cell death in Becn1 heterozygosity. In conclusion, our data first highlight a novel role of BECLIN1 in protecting against cellular ER stress independent from autophagy. These novel findings open new therapeutic avenues to intervene in this important intracellular stress response pathway with a promising impact on future AKI management.
Collapse
Affiliation(s)
- Tillmann Bork
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Camila Hernando-Erhard
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Wei Liang
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan 430064, China
| | - Zhejia Tian
- Department of Nephrology and Hypertension, Hannover Medical School, 30625 Hannover, Germany
| | - Kosuke Yamahara
- Department of Medicine IV, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Department of Medicine, Shiga University of Medical Science, Tsukinowa-cho, Otsu 520-2192, Shiga, Japan
| | - Tobias B Huber
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
11
|
Travis LB, Feldman DR, Fung C, Poynter JN, Lockley M, Frazier AL. Adolescent and Young Adult Germ Cell Tumors: Epidemiology, Genomics, Treatment, and Survivorship. J Clin Oncol 2024; 42:696-706. [PMID: 37820296 DOI: 10.1200/jco.23.01099] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/05/2023] [Accepted: 08/11/2023] [Indexed: 10/13/2023] Open
Abstract
Innovations in the care of adolescent and young adult (AYA) germ cell tumors (GCTs) are needed for one of the most common AYA cancers for which treatment has not significantly changed for several decades. Testicular GCTs (TGCTs) are the most common cancers in 15- to 39-year-old men, and ovarian GCTs (OvGCTs) are the leading gynecologic malignancies in women younger than 25 years. Excellent outcomes, even in widely metastatic disease using cisplatin-based chemotherapy, can be achieved since Einhorn and Donohue's landmark 1977 study in TGCT. However, as the severity of accompanying late effects (ototoxicity, neurotoxicity, cardiovascular disease, second malignant neoplasms, nephrotoxicity, and others) has emerged, efforts to deintensity treatment and find alternatives to cisplatin have taken on new urgency. Current innovations include the collaborative design of clinical trials that accrue GCTs across all ages and both sexes, including adolescents (previously on pediatric trials), and OvGCT (previously on gynecologic-only trials). Joint trials accrue larger sample sizes at a faster rate and therefore evaluate new approaches more rapidly. These joint trials also allow for biospecimen collection to further probe GCT etiology and underlying mechanisms of tumor growth, thus providing new therapeutic options. This AYA approach has been fostered by The Malignant Germ Cell International Consortium, which includes over 115 GCT disease experts from pediatric, gynecologic, and genitourinary oncologies in 16 countries. Trials in development incorporate, to our knowledge, for the first time, molecular risk stratification and precision oncology approaches on the basis of specific GCT biology. This collaborative AYA approach pioneering successfully in GCT could serve as a model for impactful research for other AYA cancer types.
Collapse
Affiliation(s)
- Lois B Travis
- Department of Medical Oncology, Indiana University, Indianapolis, IN
| | | | - Chunkit Fung
- J.P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY
| | - Jenny N Poynter
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | - Michelle Lockley
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - A Lindsay Frazier
- Dana Farber-Boston Children's Cancer and Blood Disorders Center, Boston, MA
| |
Collapse
|
12
|
Rossini E, Tamburello M, Abate A, Zini S, Ribaudo G, Gianoncelli A, Calza S, Valcamonico F, Suardi NR, Mirabella G, Berruti A, Sigala S. The CDK Inhibitor Dinaciclib Improves Cisplatin Response in Nonseminomatous Testicular Cancer: A Preclinical Study. Cells 2024; 13:368. [PMID: 38474332 DOI: 10.3390/cells13050368] [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: 12/13/2023] [Revised: 02/08/2024] [Accepted: 02/17/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Most patients with testicular germ cell tumors (GCTs) are treated with cisplatin (CP)-based chemotherapy. However, some of them may develop CP resistance and therefore represent a clinical challenge. Cyclin-dependent kinase 5 (CDK5) is involved in chemotherapy resistance in different types of cancer. Here, we investigated the possible role of CDK5 and other CDKs targeted by dinaciclib in nonseminoma cell models (both CP-sensitive and CP-resistant), evaluating the potential of the CDK inhibitor dinaciclib as a single/combined agent for the treatment of advanced/metastatic testicular cancer (TC). METHODS The effects of dinaciclib and CP on sensitive and resistant NT2/D1 and NCCIT cell viability and proliferation were evaluated using MTT assays and direct count methods. Flow cytometry cell-cycle analysis was performed. The protein expression was assessed via Western blotting. The in vivo experiments were conducted in zebrafish embryos xenografted with TC cells. RESULTS Among all the CDKs analyzed, CDK5 protein expression was significantly higher in CP-resistant models. Dinaciclib reduced the cell viability and proliferation in each cell model, inducing changes in cell-cycle distribution. In drug combination experiments, dinaciclib enhances the CP effect both in vitro and in the zebrafish model. CONCLUSIONS Dinaciclib, when combined with CP, could be useful for improving nonseminoma TC response to CP.
Collapse
Affiliation(s)
- Elisa Rossini
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Mariangela Tamburello
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Andrea Abate
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Silvia Zini
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Giovanni Ribaudo
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Alessandra Gianoncelli
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Stefano Calza
- Unit of Biostatistics and Bioinformatics, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Francesca Valcamonico
- Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia at ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Nazareno R Suardi
- Urology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia at ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Giuseppe Mirabella
- Urology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia at ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Alfredo Berruti
- Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia at ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Sandra Sigala
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
13
|
Fonini JS, de Araujo PHXN, D'Ambrosio PD, Salerno JVDO, Ciaralo PPD, Terra RM, Pêgo-Fernandes PM. Prolonged survival after thoracic metastasectomy in patients with nonseminomatous testicular cancer. Clinics (Sao Paulo) 2024; 79:100338. [PMID: 38359698 PMCID: PMC10877677 DOI: 10.1016/j.clinsp.2024.100338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/18/2024] [Indexed: 02/17/2024] Open
Abstract
INTRODUCTION Almost 20 % of patients with Non-Seminomatous Germinative Cell Tumors (NSGCT) will require intrathoracic metastasectomy after chemotherapy. The authors aim to determine their long-term survival rates. METHODS Retrospective study including patients with NSGCT and intrathoracic metastasis after systemic therapy from January 2011 to June 2022. Treatment outcomes and overall survival were analyzed with the Kaplan-Meier method. RESULTS Thirty-seven male patients were included with a median age of 31.8 years. Six presented with synchronous mediastinum and lung metastasis, nine had only lung, and 22 had mediastinal metastasis. Over half had retroperitoneal lymph node metastasis. Twenty-two had dissimilar pathologies, with a discordance rate of 62 %. Teratoma and embryonal carcinoma were the prevalent primary tumor types, 40.5 % each, while teratoma was predominant (70.3 %) in the metastasis group. Thoracotomy was the main surgical approach (39.2 %) followed by VATS (37.2 %), cervico-sternotomy (9.8 %), sternotomy (5.8 %), and clamshell (3.9 %). Lung resection was performed in 40.5 % of cases. Overall, 10-year survival rates were 94.3 % with no surgical-related mortality. CONCLUSION Multimodality treatment with systemic therapy followed by radical surgery offers a high cure rate to patients with intrathoracic metastatic testicular germ cell tumors.
Collapse
Affiliation(s)
- Jaqueline Schaparini Fonini
- Thoracic Surgery Department, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil.
| | - Pedro Henrique Xavier Nabuco de Araujo
- Thoracic Surgical Oncology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil; Thoracic Surgical Oncology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Paula Duarte D'Ambrosio
- Thoracic Surgery Department, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | | | - Pedro Prosperi Desenzi Ciaralo
- Thoracic Surgery Department, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Ricardo Mingarini Terra
- Thoracic Surgery Department, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil; Thoracic Surgery Department, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | - Paulo Manuel Pêgo-Fernandes
- Thoracic Surgery Department, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| |
Collapse
|
14
|
van der Meer DJ, Karim-Kos HE, Elzevier HW, Dinkelman-Smit M, Kerst JM, Atema V, Lehmann V, Husson O, van der Graaf WTA. The increasing burden of testicular seminomas and non-seminomas in adolescents and young adults (AYAs): incidence, treatment, disease-specific survival and mortality trends in the Netherlands between 1989 and 2019. ESMO Open 2024; 9:102231. [PMID: 38244349 PMCID: PMC10937200 DOI: 10.1016/j.esmoop.2023.102231] [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: 08/21/2023] [Revised: 11/11/2023] [Accepted: 12/21/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Testicular cancer incidence among adolescents and young adults (AYAs, aged 18-39 years at diagnosis) is increasing worldwide and most patients will survive the initial disease. Still, detailed epidemiological information about testicular cancer among AYAs is scarce. This study aimed to provide a detailed overview of testicular cancer trends in incidence, treatment, long-term relative survival and mortality by histological subtype among AYAs diagnosed in the Netherlands between 1989 and 2019. MATERIALS AND METHODS Data of all malignant testicular cancers (ICD-code C62) were extracted from the Netherlands Cancer Registry. Mortality data were retrieved from Statistics Netherlands. European age-standardized incidence and mortality rates with average annual percentage change statistics and relative survival estimates up to 20 years of follow-up were calculated. RESULTS A total of 12 528 testicular cancers were diagnosed between 1989 and 2019. Comparing 1989-1999 to 2010-2019, the incidence increased from 4.4 to 11.4 for seminomas and from 5.7 to 11.1 per 100 000 person-years for non-seminomas. Rising trends were most prominent for localized disease. Radiotherapy use in localized testicular seminomas declined from 78% in 1989-1993 to 5% in 2015-2019. Meanwhile, there was a slight increase in chemotherapy use. Most AYAs with localized seminomas and non-seminomas received active surveillance only (>80%). Overall, relative survival estimates remained well above 90% even at 20 years of follow-up for both seminomas and non-seminomas. Mortality rates declined from 0.5 to 0.4 per 100 000 person-years between 1989-1999 and 2010-2019. CONCLUSIONS The incidence of seminoma and non-seminoma testicular cancers significantly increased in AYAs in the Netherlands between 1989 and 2019. There was a shift towards less-aggressive treatment regimens without negative survival effects. Relative survival estimates remained well above 90% at 20 years of follow-up in most cases. Testicular cancer mortality was already low, but has improved further over time, which makes survivorship care an important issue for these young adults.
Collapse
Affiliation(s)
- D J van der Meer
- Department of Medical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam; Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam.
| | - H E Karim-Kos
- Princess Máxima Center for Pediatric Oncology, Utrecht; Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht
| | - H W Elzevier
- Department of Urology and Medical Decision Making, Leiden University Medical Centre, Leiden
| | - M Dinkelman-Smit
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam
| | - J M Kerst
- Department of Medical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam
| | - V Atema
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht
| | - V Lehmann
- Department of Medical Psychology, Cancer Center Amsterdam, Amsterdam University Medical Center, Amsterdam; Cancer Center Amsterdam (CCA), Amsterdam
| | - O Husson
- Department of Medical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam; Department of Surgical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - W T A van der Graaf
- Department of Medical Oncology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam; Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam
| |
Collapse
|
15
|
Schütte W, Gütz S, Nehls W, Blum TG, Brückl W, Buttmann-Schweiger N, Büttner R, Christopoulos P, Delis S, Deppermann KM, Dickgreber N, Eberhardt W, Eggeling S, Fleckenstein J, Flentje M, Frost N, Griesinger F, Grohé C, Gröschel A, Guckenberger M, Hecker E, Hoffmann H, Huber RM, Junker K, Kauczor HU, Kollmeier J, Kraywinkel K, Krüger M, Kugler C, Möller M, Nestle U, Passlick B, Pfannschmidt J, Reck M, Reinmuth N, Rübe C, Scheubel R, Schumann C, Sebastian M, Serke M, Stoelben E, Stuschke M, Thomas M, Tufman A, Vordermark D, Waller C, Wolf J, Wolf M, Wormanns D. [Prevention, Diagnosis, Therapy, and Follow-up of Lung Cancer - Interdisciplinary Guideline of the German Respiratory Society and the German Cancer Society - Abridged Version]. Pneumologie 2023; 77:671-813. [PMID: 37884003 DOI: 10.1055/a-2029-0134] [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: 10/28/2023]
Abstract
The current S3 Lung Cancer Guidelines are edited with fundamental changes to the previous edition based on the dynamic influx of information to this field:The recommendations include de novo a mandatory case presentation for all patients with lung cancer in a multidisciplinary tumor board before initiation of treatment, furthermore CT-Screening for asymptomatic patients at risk (after federal approval), recommendations for incidental lung nodule management , molecular testing of all NSCLC independent of subtypes, EGFR-mutations in resectable early stage lung cancer in relapsed or recurrent disease, adjuvant TKI-therapy in the presence of common EGFR-mutations, adjuvant consolidation treatment with checkpoint inhibitors in resected lung cancer with PD-L1 ≥ 50%, obligatory evaluation of PD-L1-status, consolidation treatment with checkpoint inhibition after radiochemotherapy in patients with PD-L1-pos. tumor, adjuvant consolidation treatment with checkpoint inhibition in patients withPD-L1 ≥ 50% stage IIIA and treatment options in PD-L1 ≥ 50% tumors independent of PD-L1status and targeted therapy and treatment option immune chemotherapy in first line SCLC patients.Based on the current dynamic status of information in this field and the turnaround time required to implement new options, a transformation to a "living guideline" was proposed.
Collapse
Affiliation(s)
- Wolfgang Schütte
- Klinik für Innere Medizin II, Krankenhaus Martha Maria Halle-Dölau, Halle (Saale)
| | - Sylvia Gütz
- St. Elisabeth-Krankenhaus Leipzig, Abteilung für Innere Medizin I, Leipzig
| | - Wiebke Nehls
- Klinik für Palliativmedizin und Geriatrie, Helios Klinikum Emil von Behring
| | - Torsten Gerriet Blum
- Helios Klinikum Emil von Behring, Klinik für Pneumologie, Lungenklinik Heckeshorn, Berlin
| | - Wolfgang Brückl
- Klinik für Innere Medizin 3, Schwerpunkt Pneumologie, Klinikum Nürnberg Nord
| | | | - Reinhard Büttner
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Uniklinik Köln, Berlin
| | | | - Sandra Delis
- Helios Klinikum Emil von Behring, Klinik für Pneumologie, Lungenklinik Heckeshorn, Berlin
| | | | - Nikolas Dickgreber
- Klinik für Pneumologie, Thoraxonkologie und Beatmungsmedizin, Klinikum Rheine
| | | | - Stephan Eggeling
- Vivantes Netzwerk für Gesundheit, Klinikum Neukölln, Klinik für Thoraxchirurgie, Berlin
| | - Jochen Fleckenstein
- Klinik für Strahlentherapie und Radioonkologie, Universitätsklinikum des Saarlandes und Medizinische Fakultät der Universität des Saarlandes, Homburg
| | - Michael Flentje
- Klinik und Poliklinik für Strahlentherapie, Universitätsklinikum Würzburg, Würzburg
| | - Nikolaj Frost
- Medizinische Klinik mit Schwerpunkt Infektiologie/Pneumologie, Charite Universitätsmedizin Berlin, Berlin
| | - Frank Griesinger
- Klinik für Hämatologie und Onkologie, Pius-Hospital Oldenburg, Oldenburg
| | | | - Andreas Gröschel
- Klinik für Pneumologie und Beatmungsmedizin, Clemenshospital, Münster
| | | | | | - Hans Hoffmann
- Klinikum Rechts der Isar, TU München, Sektion für Thoraxchirurgie, München
| | - Rudolf M Huber
- Medizinische Klinik und Poliklinik V, Thorakale Onkologie, LMU Klinikum Munchen
| | - Klaus Junker
- Klinikum Oststadt Bremen, Institut für Pathologie, Bremen
| | - Hans-Ulrich Kauczor
- Klinikum der Universität Heidelberg, Abteilung Diagnostische Radiologie, Heidelberg
| | - Jens Kollmeier
- Helios Klinikum Emil von Behring, Klinik für Pneumologie, Lungenklinik Heckeshorn, Berlin
| | | | - Marcus Krüger
- Klinik für Thoraxchirurgie, Krankenhaus Martha-Maria Halle-Dölau, Halle-Dölau
| | | | - Miriam Möller
- Krankenhaus Martha-Maria Halle-Dölau, Klinik für Innere Medizin II, Halle-Dölau
| | - Ursula Nestle
- Kliniken Maria Hilf, Klinik für Strahlentherapie, Mönchengladbach
| | | | - Joachim Pfannschmidt
- Klinik für Thoraxchirurgie, Lungenklinik Heckeshorn, Helios Klinikum Emil von Behring, Berlin
| | - Martin Reck
- Lungeclinic Grosshansdorf, Pneumologisch-onkologische Abteilung, Grosshansdorf
| | - Niels Reinmuth
- Klinik für Pneumologie, Thorakale Onkologie, Asklepios Lungenklinik Gauting, Gauting
| | - Christian Rübe
- Klinik für Strahlentherapie und Radioonkologie, Universitätsklinikum des Saarlandes, Homburg/Saar, Homburg
| | | | | | - Martin Sebastian
- Medizinische Klinik II, Universitätsklinikum Frankfurt, Frankfurt
| | - Monika Serke
- Zentrum für Pneumologie und Thoraxchirurgie, Lungenklinik Hemer, Hemer
| | | | - Martin Stuschke
- Klinik und Poliklinik für Strahlentherapie, Universitätsklinikum Essen, Essen
| | - Michael Thomas
- Thoraxklinik am Univ.-Klinikum Heidelberg, Thorakale Onkologie, Heidelberg
| | - Amanda Tufman
- Medizinische Klinik und Poliklinik V, Thorakale Onkologie, LMU Klinikum München
| | - Dirk Vordermark
- Universitätsklinik und Poliklinik für Strahlentherapie, Universitätsklinikum Halle, Halle
| | - Cornelius Waller
- Klinik für Innere Medizin I, Universitätsklinikum Freiburg, Freiburg
| | | | - Martin Wolf
- Klinikum Kassel, Klinik für Onkologie und Hämatologie, Kassel
| | - Dag Wormanns
- Evangelische Lungenklinik, Radiologisches Institut, Berlin
| |
Collapse
|
16
|
Bailey S, Ferraresso M, Alonso-Crisostomo L, Ward D, Smith S, Nicholson JC, Saini H, Enright AJ, Scarpini CG, Coleman N, Murray MJ. Targeting oncogenic microRNAs from the miR-371~373 and miR-302/367 clusters in malignant germ cell tumours causes growth inhibition through cell cycle disruption. Br J Cancer 2023; 129:1451-1461. [PMID: 37789102 PMCID: PMC10628203 DOI: 10.1038/s41416-023-02453-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 09/13/2023] [Accepted: 09/21/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND MiR-371~373 and miR-302/367 cluster over-expression occurs in all malignant germ cell tumours (GCTs), regardless of age (paediatric/adult), site (gonadal/extragonadal), or subtype [seminoma, yolk sac tumour (YST), embryonal carcinoma (EC)]. Six of eight microRNAs from these clusters contain the seed sequence 'AAGUGC', determining mRNA targeting. Here we sought to identify the significance of these observations by targeting these microRNAs functionally. METHODS We targeted miR-371~373 and/or miR-302/367 clusters in malignant GCT cell lines, using CRISPR-Cas9, gapmer primary miR-302/367 transcript inhibition, and peptide nucleic acid (PNA) or locked nucleic acid (LNA)-DNA inhibition targeting miR-302a-d-3p, and undertook relevant functional assays. RESULTS MiR-302/367 cluster microRNAs made the largest contribution to AAGUGC seed abundance in malignant GCT cells, regardless of subtype (seminoma/YST/EC). Following the unsuccessful use of CRISPR-Cas9, gapmer, and PNA systems, LNA-DNA-based targeting resulted in growth inhibition in seminoma and YST cells. This was associated with the de-repression of multiple mRNAs targeted by AAGUGC seed-containing microRNAs, with pathway analysis confirming predominant disruption of Rho-GTPase signalling, vesicle organisation/transport, and cell cycle regulation, findings corroborated in clinical samples. Further LNA-DNA inhibitor studies confirmed direct cell cycle effects, with an increase of cells in G0/G1-phase and a decrease in S-phase. CONCLUSION Targeting of specific miR-371~373 and miR-302/367 microRNAs in malignant GCTs demonstrated their functional significance, with growth inhibition mediated through cell cycle disruption.
Collapse
Affiliation(s)
- Shivani Bailey
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
- Department of Paediatric Haematology and Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Marta Ferraresso
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | | | - Dawn Ward
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Stephen Smith
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - James C Nicholson
- Department of Paediatric Haematology and Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
- Department of Paediatrics, University of Cambridge, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK
| | - Harpreet Saini
- EMBL-European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Anton J Enright
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Cinzia G Scarpini
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Nicholas Coleman
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK.
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK.
| | - Matthew J Murray
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK.
- Department of Paediatric Haematology and Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
17
|
Wang Y, Tang S, Li L, Sun C, Liu Y, Zhao Y. Depletion of circPDSS1 inhibits ITGA11 production to confer cisplatin sensitivity through miR-515-5p in gastric cancer. J Chemother 2023; 35:514-526. [PMID: 36484486 DOI: 10.1080/1120009x.2022.2151702] [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: 07/05/2022] [Revised: 11/04/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022]
Abstract
Chemoresistance limits cisplatin (DDP)-mediated treatment for gastric cancer (GC). Circular RNA (circRNA) acts an important role in chemoresistance. However, the underlying mechanism of circPDSS1 regulating DDP sensitivity in GC remains unclear. The expression patterns of circPDSS1, miR-515-5p and integrin subunit alpha 11 (ITGA11) were analyzed by qRT-PCR. Protein expression was checked by Western blotting analysis. Cell viability was investigated by 3-(4,5-dimethylthazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cell proliferation was evaluated by colony formation assay and 5-ethynyl-2'-deoxyuridine (EdU) assay. The analysis of cell apoptosis, migration and invasion was performed by flow cytometry analysis and transwell assays. Dual-luciferase reporter assay and RNA immunoprecipitation assay were conducted to identify the associations among circPDSS1, miR-515-5p and ITGA11. In vivo assay was implemented using a xenograft mouse model assay. CircPDSS1 and ITGA11 expression were significantly upregulated, whereas miR-515-5p was downregulated in DDP-resistant GC tissues and cells in comparison with controls. CircPDSS1 depletion reduced DDP resistance, cell proliferation, migration and invasion but induced cell apoptosis in DDP-resistant GC cells. CircPDSS1 directly bound to miR-515-5p. CircPDSS1-mediated actions were dependent on the regulation of miR-515-5p. Besides, miR-515-5p was associated with ITGA11, and circPDSS1 regulated ITGA11 expression by binding to miR-515-5p. Overexpression of miR-515-5p improved DDP sensitivity owing to the downregulation of ITGA11. Further, circPDSS1 mediated DDP sensitivity by regulating miR-515-5p and ITGA11 in vivo. CircPDSS1 conferred DDP resistance through the miR-515-5p/ITGA11 axis in GC cells.
Collapse
Affiliation(s)
- Yongsen Wang
- Department of Digestive Medicine, Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuting Tang
- Department of Internal Medicine, Wendeng Osteopathic Hospital of Shandong Province, Weihai, China
| | - Lingling Li
- Clinical College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cheng Sun
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yaru Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yujie Zhao
- Department of Digestive Medicine, Hospital Affiliated to Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
18
|
Mapuskar KA, Pulliam CF, Zepeda-Orozco D, Griffin BR, Furqan M, Spitz DR, Allen BG. Redox Regulation of Nrf2 in Cisplatin-Induced Kidney Injury. Antioxidants (Basel) 2023; 12:1728. [PMID: 37760031 PMCID: PMC10525889 DOI: 10.3390/antiox12091728] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/30/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Cisplatin, a potent chemotherapeutic agent, is marred by severe nephrotoxicity that is governed by mechanisms involving oxidative stress, inflammation, and apoptosis pathways. The transcription factor Nrf2, pivotal in cellular defense against oxidative stress and inflammation, is the master regulator of the antioxidant response, upregulating antioxidants and cytoprotective genes under oxidative stress. This review discusses the mechanisms underlying chemotherapy-induced kidney injury, focusing on the role of Nrf2 in cancer therapy and its redox regulation in cisplatin-induced kidney injury. We also explore Nrf2's signaling pathways, post-translational modifications, and its involvement in autophagy, as well as examine redox-based strategies for modulating Nrf2 in cisplatin-induced kidney injury while considering the limitations and potential off-target effects of Nrf2 modulation. Understanding the redox regulation of Nrf2 in cisplatin-induced kidney injury holds significant promise for developing novel therapeutic interventions. This knowledge could provide valuable insights into potential strategies for mitigating the nephrotoxicity associated with cisplatin, ultimately enhancing the safety and efficacy of cancer treatment.
Collapse
Affiliation(s)
- Kranti A. Mapuskar
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| | - Casey F. Pulliam
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| | - Diana Zepeda-Orozco
- Pediatric Nephrology and Hypertension at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| | - Benjamin R. Griffin
- Division of Nephrology, The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
- Department of Internal Medicine, The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Muhammad Furqan
- Department of Internal Medicine, The University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| | - Bryan G. Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, USA
| |
Collapse
|
19
|
Wang Z, Li B, Xing J, Gong Z, Xu A, Wang Z. Causes of death after testicular cancer diagnosis: a US population-based analysis. BMC Urol 2023; 23:144. [PMID: 37660082 PMCID: PMC10475185 DOI: 10.1186/s12894-023-01309-3] [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: 07/09/2022] [Accepted: 08/10/2023] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND After the introduction of cisplatin-based chemotherapy, the survival time of testicular cancer (TC) patients has improved dramatically. However, the overall risk of death in patients with TC remains significantly higher than in the general population. The aim of this study was to assess and quantify the causes of death after TC diagnosis. METHOD In total, 44,975 men with TC in the United States diagnosed and registered by the Surveillance, Epidemiology, and End Results (SEER) database during 2000 to 2018 were studied. In this study, standardized mortality rates (SMRs) were calculated for each cause of death in TC individuals and further analyzed in strata according to age and race. RESULT Of the included participants, 3,573 (7.94%) died during the follow-up period. The greatest proportion of deaths (38.20%) occurred within 1 to 5 years after diagnosis. Most deaths occurred from TC itself and other cancers. For non-malignant conditions, the most common causes of death within 1 years after diagnosis were accidents and adverse effects (53, 4.75%) followed by diseases of heart (45, 4.04%). However, > 1 years after diagnosis, the most common noncancer causes of death were heart diseases. Results of stratified analysis show that non-Hispanic White TC participants have a lower SMR (0.68, 95% CI, 33.39-38.67) from Cerebrovascular Diseases than the general U.S. POPULATION CONCLUSIONS Although TC remains the most common cause of death after TC diagnosis, other non-TC causes of death represent a significant number of deaths among TC men. These findings help TC survivors understand the various health risks that may occur at different follow-up periods.
Collapse
Affiliation(s)
- Zhongyuan Wang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, 210029, Jiangsu Province, China
| | - Baochao Li
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, 210029, Jiangsu Province, China
| | - Jiajun Xing
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, 210029, Jiangsu Province, China
| | - Zixuan Gong
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, 210029, Jiangsu Province, China
| | - Aiming Xu
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, 210029, Jiangsu Province, China.
| | - Zengjun Wang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
20
|
Garcia SL, Lauritsen J, Christiansen BK, Hansen IF, Bandak M, Dalgaard MD, Daugaard G, Gupta R. Predicting Hearing Loss in Testicular Cancer Patients after Cisplatin-Based Chemotherapy. Cancers (Basel) 2023; 15:3923. [PMID: 37568739 PMCID: PMC10417151 DOI: 10.3390/cancers15153923] [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: 06/19/2023] [Revised: 07/25/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
Testicular cancer is predominantly curable, but the long-term side effects of chemotherapy have a severe impact on life quality. In this research study, we focus on hearing loss as a part of overall chemotherapy-induced ototoxicity. This is a unique approach where we combine clinical data from the acclaimed nationwide Danish Testicular Cancer (DaTeCa)-Late database. Clinical and genetic data on 433 patients were collected from hospital files in October 2014. Hearing loss was classified according to the FACT/GOG-Ntx-11 version 4 self-reported Ntx6. Machine learning models combining a genome-wide association study within a nested cross-validated logistic regression were applied to identify patients at high risk of hearing loss. The model comprising clinical and genetic data identified 67% of the patients with hearing loss; however, this was with a false discovery rate of 49%. For the non-affected patients, the model identified 66% of the patients with a false omission rate of 19%. An area under the receiver operating characteristic (ROC-AUC) curve of 0.73 (95% CI, 0.71-0.74) was obtained, and the model suggests genes SOD2 and MGST3 as important in improving prediction over the clinical-only model with a ROC-AUC of 0.66 (95% CI, 0.65-0.66). Such prediction models may be used to allow earlier detection and prevention of hearing loss. We suggest a possible biological mechanism for cisplatin-induced hearing loss development. On confirmation in larger studies, such models can help balance treatment in clinical practice.
Collapse
Affiliation(s)
- Sara L. Garcia
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; (S.L.G.); (R.G.)
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 1958 Copenhagen, Denmark
| | - Jakob Lauritsen
- Department of Oncology, Copenhagen University Hospital, 2730 Copenhagen, Denmark
| | - Bernadette K. Christiansen
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; (S.L.G.); (R.G.)
| | - Ida F. Hansen
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; (S.L.G.); (R.G.)
| | - Mikkel Bandak
- Department of Oncology, Copenhagen University Hospital, 2730 Copenhagen, Denmark
| | - Marlene D. Dalgaard
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; (S.L.G.); (R.G.)
| | - Gedske Daugaard
- Department of Oncology, Copenhagen University Hospital, 2730 Copenhagen, Denmark
| | - Ramneek Gupta
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; (S.L.G.); (R.G.)
- Department of Computational Biology, Novo Nordisk Research Centre Oxford, Oxford OX3 7FZ, UK
| |
Collapse
|
21
|
Wu P, Yang Y, Yu Z, Zhao L, Feng S. Clinical Features and Survival Outcomes in Children and Adolescents With Malignant Mediastinal Germ Cell Tumors Based on Surveillance, Epidemiology, and End Results Database Analysis. J Surg Res 2023; 288:362-371. [PMID: 37062232 DOI: 10.1016/j.jss.2023.03.018] [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: 09/27/2022] [Revised: 02/28/2023] [Accepted: 03/16/2023] [Indexed: 04/18/2023]
Abstract
INTRODUCTION The purpose of this study was to perform a population-based investigation to assess the disease characteristics and prognosis of children and adolescents with malignant mediastinal germ cell tumors (MMGCT). METHODS Data on the demographics, treatment, and survival outcomes of children and adolescents with MMGCT from January 1, 2000 to December 31, 2018 were obtained. To compare survival curves, the log-rank test was employed. The generation of survival curves based on different parameters was done using Kaplan-Meier estimations. Cox proportional hazards regression was performed to determine the variables linked to disease-specific survival. RESULTS The selection criteria were met by 152 MMGCT patients, 130 of whom were male. Fifty three cases of mixed germ cell tumors (GCTs), 41 cases of malignant teratomas, 26 cases of yolk sac tumors, 14 cases of seminoma, 13 cases of choriocarcinomas, and five cases of embryonal carcinoma were reported. Overall survival at 3 and 5 y for all patients was 63.1% and 61.2%, respectively. Malignant teratoma, yolk sac tumors, and mixed GCTs in children and adolescents had comparable survival rates, while those with choriocarcinoma and embryonal carcinoma showed the worst prognosis. Embryonal carcinoma, malignant teratoma, mixed GCTs, and choriocarcinoma were found as risk factors by multivariate Cox proportional hazards analysis. In contrast, surgery and younger age were protective factors. However, chemotherapy alone showed no survival benefits. CONCLUSIONS Our population-based evidence showed that MMGCT had worse prognosis in older children and adolescents. Choriocarcinomas and embryonal carcinomas had the worst prognosis. Surgery can prolong survival time. Chemotherapy and radiotherapy were not associated with improved prognosis.
Collapse
Affiliation(s)
- Peng Wu
- Department of Pediatric Surgery, Northwest Women and Children's Hospital, Xi'an, China
| | - Yicheng Yang
- Department of Urology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Zhechen Yu
- Department of Urology, Children's Hospital of Soochow University, Suzhou, Jiangsu, P.R. China
| | - Lingling Zhao
- Department of Pathology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Shaoguang Feng
- Department of Pediatric Surgery, Hangzhou Children's Hospital, Hangzhou, China.
| |
Collapse
|
22
|
Zhang Y, Bao S, Wang D, Lu W, Xu S, Zhou W, Wang X, Xu X, Ding X, Zhao S. Downregulation of KLF10 contributes to the regeneration of survived renal tubular cells in cisplatin-induced acute kidney injury via ZBTB7A-KLF10-PTEN axis. Cell Death Discov 2023; 9:82. [PMID: 36878898 PMCID: PMC9988960 DOI: 10.1038/s41420-023-01381-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/08/2023] Open
Abstract
Acute kidney injury (AKI) is a common clinical dysfunction with complicated pathophysiology and limited therapeutic methods. Renal tubular injury and the following regeneration process play a vital role in the course of AKI, but the underlining molecular mechanism remains unclear. In this study, network-based analysis of online transcriptional data of human kidney found that KLF10 was closely related to renal function, tubular injury and regeneration in various renal diseases. Three classical mouse models confirmed the downregulation of KLF10 in AKI and its correlation with tubular regeneration and AKI outcome. The 3D renal tubular model in vitro and fluorescent visualization system of cellular proliferation were constructed to show that KLF10 declined in survived cells but increased during tubular formation or conquering proliferative impediment. Furthermore, overexpression of KLF10 significantly inhibited, whereas knockdown of KLF10 extremely promoted the capacity of proliferation, injury repairing and lumen-formation of renal tubular cells. In mechanism, PTEN/AKT pathway were validated as the downstream of KLF10 and participated in its regulation of tubular regeneration. By adopting proteomic mass spectrum and dual-luciferase reporter assay, ZBTB7A were found to be the upstream transcription factor of KLF10. Our findings suggest that downregulation of KLF10 positively contributed to tubular regeneration in cisplatin induced acute kidney injury via ZBTB7A-KLF10-PTEN axis, which gives insight into the novel therapeutic and diagnostical target of AKI.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Siyu Bao
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Daxi Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Lu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sujuan Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weiran Zhou
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyan Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xialian Xu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China. .,Shanghai Medical Center of Kidney Disease, Shanghai, China. .,Kidney and Dialysis Institute of Shanghai, Shanghai, China. .,Kidney and Blood Purification Key Laboratory of Shanghai, Shanghai, China.
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China. .,Shanghai Medical Center of Kidney Disease, Shanghai, China. .,Kidney and Dialysis Institute of Shanghai, Shanghai, China. .,Kidney and Blood Purification Key Laboratory of Shanghai, Shanghai, China.
| | - Shuan Zhao
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China. .,Shanghai Medical Center of Kidney Disease, Shanghai, China. .,Kidney and Dialysis Institute of Shanghai, Shanghai, China. .,Kidney and Blood Purification Key Laboratory of Shanghai, Shanghai, China.
| |
Collapse
|
23
|
Tanuma K, Kawai K, Nitta S, Shiga M, Kawahara T, Negoro H, Onozawa M, Inoue T, Nishiyama H, Miyazaki J. Improved survival of poor-risk non-seminomatous germ cell tumor patients: real-world data from a single institute in Japan. Jpn J Clin Oncol 2023; 53:74-79. [PMID: 36151048 DOI: 10.1093/jjco/hyac151] [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: 08/03/2022] [Accepted: 08/31/2022] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVES The International Germ Cell Cancer Collaborative Group Update Consortium showed the improved survival of patients with a non-seminomatous germ cell tumor. We updated the survival data of the non-seminomatous germ cell tumor patients treated at our hospital. PATIENTS AND METHODS We analyzed the outcomes of 138 patients treated in 1981-2018. We compared the survival of the patients treated in the early (1981-99) and later (2000-18) periods and determined the groups' progression-free survival and overall survival using the Kaplan-Meier method. We used a web-based application of the International Germ Cell Cancer Collaborative Group Update model to calculate each patient's predicted 3-year progression-free survival. RESULTS The 5-year progression-free survival rates of the good, intermediate and poor prognosis groups were 91, 83 and 64%, and their 5-year overall survival rates were 97, 89 and 82%, respectively. There were no significant differences in the progression-free survival or overall survival of the good and intermediate prognosis groups by treatment year. The 5-year progression-free survival of the poor prognosis group was almost identical in both treatment year (60 and 65%, respectively). By contrast, the 5-year overall survival in the later period (85%) was higher than that in the early period (70%). The median-predicted 3-year progression-free survival rates of the good, intermediate and poor prognosis groups were 92, 83 and 51% (P < 0.01), respectively. The concordance index for the good, intermediate and poor prognosis groups were 0.56, 0.79 and 0.67, respectively. CONCLUSION The survival of our poor prognosis non-seminomatous germ cell tumor patients improved over time. The 5-year overall survival of patients treated in 2000-18 reached 85%.
Collapse
Affiliation(s)
- Kozaburo Tanuma
- Department of Urology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Koji Kawai
- Department of Urology, International University of Health and Welfare Narita Hospital, Chiba, Japan
| | - Satoshi Nitta
- Department of Urology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Masanobu Shiga
- Department of Urology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Takashi Kawahara
- Department of Urology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Hiromitsu Negoro
- Department of Urology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Mizuki Onozawa
- Department of Urology, International University of Health and Welfare Narita Hospital, Chiba, Japan
| | - Takamitsu Inoue
- Department of Urology, International University of Health and Welfare Narita Hospital, Chiba, Japan
| | - Hiroyuki Nishiyama
- Department of Urology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Jun Miyazaki
- Department of Urology, International University of Health and Welfare Narita Hospital, Chiba, Japan
| |
Collapse
|
24
|
Chovanec M, Adra N, Abu Zaid M, Abonour R, Einhorn L. High-dose chemotherapy for relapsed testicular germ cell tumours. Nat Rev Urol 2022; 20:217-225. [PMID: 36477219 DOI: 10.1038/s41585-022-00683-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
Relapsed testicular germ cell tumours (GCTs) might be cured with salvage chemotherapy. Accepted salvage treatment is conventional-dose chemotherapy (CDCT) or high-dose chemotherapy (HDCT). HDCT with peripheral blood stem cell transplant might produce a higher number of durable responses than CDCT. We discuss studies reporting on outcomes of salvage HDCT in relapsed GCTs. The most reproducible results were achieved with HDCT with two cycles of etoposide and carboplatin or three cycles of the paclitaxel, ifosfamide, carboplatin and etoposide regime. Using these two regimens, sustained cure rates of 50-66% were reported in phase I, phase II and retrospective studies published in the past two decades. Cure rates in patients with cisplatin-resistant disease are between 30% and 45%. Two phase III randomized studies were conducted with certain limitations and were unsuccessful in showing a survival benefit of HDCT. Thus, salvage treatment remains a controversial topic. Salvage HDCT with peripheral blood stem cell transplant and CDCT are two recommended treatment options for relapsed GCTs. Consistently reported cure rates from phase I, phase II and large retrospective studies support the use of HDCT in the hands of an experienced team of oncologists.
Collapse
|
25
|
Abstract
Testicular cancer is a curable cancer. The success of physicians in curing the disease is underpinned by multidisciplinary advances. Cisplatin-based combination chemotherapy and the refinement of post-chemotherapy surgical procedures and diagnostic strategies have greatly improved long term survival in most patients. Despite such excellent outcomes, several controversial dilemmas exist in the approaches to clinical stage I disease, salvage chemotherapy, post-chemotherapy surgical procedures, and implementing innovative imaging studies. Relapse after salvage chemotherapy has a poor prognosis and the optimal treatment is not apparent. Recent research has provided insight into the molecular mechanisms underlying cisplatin resistance. Phase 2 studies with targeted agents have failed to show adequate efficacy; however, our understanding of cisplatin resistant disease is rapidly expanding. This review summarizes recent advances and discusses relevant issues in the biology and management of testicular cancer.
Collapse
Affiliation(s)
- Michal Chovanec
- 2nd Department of Oncology, Faculty of Medicine, Comenius University, National Cancer Institute, Bratislava, Slovakia
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Lifespan Academic Medical Center, Providence, RI, USA
| |
Collapse
|
26
|
Timmerman DM, Eleveld TF, Sriram S, Dorssers LC, Gillis AJ, Schmidtova S, Kalavska K, van de Werken HJ, Oing C, Honecker F, Mego M, Looijenga LH. Chromosome 3p25.3 Gain Is Associated With Cisplatin Resistance and Is an Independent Predictor of Poor Outcome in Male Malignant Germ Cell Tumors. J Clin Oncol 2022; 40:3077-3087. [PMID: 35442716 PMCID: PMC9462533 DOI: 10.1200/jco.21.02809] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/24/2022] [Accepted: 03/14/2022] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Cisplatin is the main systemic treatment modality for male type II germ cell tumors (GCTs). Although generally very effective, 5%-10% of patients suffer from cisplatin-resistant disease. Identification of the driving mechanisms of resistance will enable improved risk stratification and development of alternative treatments. METHODS We developed and characterized cisplatin-resistant GCT cell line models and compared their molecular characteristics with patient samples with cisplatin resistance and/or a poor clinical outcome. Subsequently, the association between the overlapping genetic features and clinical data was assessed. Finally, we used Cox regression to determine the prognostic relevance of these features within the currently used risk classification. RESULTS Gain of chromosome 3p25.3 was detected in all cisplatin-resistant cell lines, and copy number of this region correlated with the level of resistance (R = 0.96, P = 1.5e-04). Gain of this region was detected at low frequencies in primary tumors and at higher frequencies in relapsed and/or cisplatin-resistant tumors. Chromosome 3p25.3 gain was associated with shorter progression-free survival and overall survival, with the strongest association observed in nonseminomas excluding pure teratomas. 3p25.3 gain was more frequently observed in tumors with yolk sac tumor histology and predicted adverse outcome independent of the International Germ Cell Cancer Collaborative Group risk classification and the presence of TP53/MDM2 alterations. CONCLUSION On the basis of both in vitro analyses and clinical data, we found 3p25.3 to be strongly associated with cisplatin resistance and poor clinical outcome in male type II GCTs. Using genomic profiling, 3p25.3 status could help to improve risk stratification in male patients with type II GCT. Further characterization of this locus and underlying mechanisms of resistance is warranted to guide development of novel treatment approaches for cisplatin-resistant disease.
Collapse
Affiliation(s)
| | - Thomas F. Eleveld
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Sruthi Sriram
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Lambert C.J. Dorssers
- Department of Pathology, Lab for Exp Patho-Oncology (LEPO), Erasmus MC-University Medical Center Rotterdam, Cancer Institute, Rotterdam, the Netherlands
| | - Ad J.M. Gillis
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Silvia Schmidtova
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Translational Research Unit, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Katarina Kalavska
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Translational Research Unit, Faculty of Medicine, Comenius University, Bratislava, Slovakia
- Second Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Harmen J.G. van de Werken
- Cancer Computational Biology Center, Department of Urology & Department of Immunology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Christoph Oing
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCs4, University Cancer Center Hamburg, University Medical Center Eppendorf, Hamburg, Germany
| | - Friedemann Honecker
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
- Tumor- and Breast Center ZeTuP, Sankt Gallen, Switzerland
| | - Michal Mego
- Cancer Research Institute, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- Translational Research Unit, Faculty of Medicine, Comenius University, Bratislava, Slovakia
- Second Department of Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | | |
Collapse
|
27
|
Znaor A, Skakkebaek NE, Rajpert-De Meyts E, Kuliš T, Laversanne M, Gurney J, Sarfati D, McGlynn KA, Bray F. Global patterns in testicular cancer incidence and mortality in 2020. Int J Cancer 2022; 151:692-698. [PMID: 35277970 DOI: 10.1002/ijc.33999] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 11/09/2022]
Abstract
With 74 500 new cases worldwide in 2020, testicular cancer ranks as the 20th leading cancer type, but is the most common cancer in young men of European ancestry. While testicular cancer incidence has been rising in many populations, mortality trends, at least those in high-income settings, have been in decline since the 1970s following the introduction of platinum-based chemotherapy. To examine current incidence and mortality patterns, we extracted the new cases of, and deaths from cancers of the testis from the GLOBOCAN 2020 database. In 2020, testicular cancer was the most common cancer in men aged 15 to 44 in 62 countries worldwide. Incidence rates were highest in West-, North- and South-Europe and Oceania (age-standardised rate, ASR ≥7/100 000), followed by North America (5.6/100 000 and lowest (<2/100 000) in Asia and Africa. The mortality rates were highest in Central and South America (0.84 and 0.54 per 100 000, respectively), followed by Eastern and Southern Europe, and Western and Southern Africa. The lowest mortality rates were in Northern Europe, Northern Africa and Eastern Asia (0.16, 0.14, 0.9 per 100 000, respectively). At the country level, incidence rates varied over 100-fold, from 10/100 000 in Norway, Slovenia, Denmark and Germany to ≤0.10/100 000 in Gambia, Guinea, Liberia, Lesotho. Mortality rates were highest in Fiji, Argentina and Mexico. Our results indicate a higher mortality burden in countries undergoing economic transitions and reinforce the need for more equitable access to testicular cancer diagnosis and treatment globally.
Collapse
Affiliation(s)
- Ariana Znaor
- Section of Cancer Surveillance, International Agency for Research on Cancer, Lyon, France
| | - Niels Erik Skakkebaek
- Department of Growth & Reproduction, Copenhagen University Hospital (Ringshospitalet), Copenhagen, Denmark
| | - Ewa Rajpert-De Meyts
- Department of Growth & Reproduction, Copenhagen University Hospital (Ringshospitalet), Copenhagen, Denmark
| | - Tomislav Kuliš
- Department of Urology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Mathieu Laversanne
- Section of Cancer Surveillance, International Agency for Research on Cancer, Lyon, France
| | - Jason Gurney
- Department of Urology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Diana Sarfati
- Department of Public Health, University of Otago, Wellington, New Zealand
| | - Katherine A McGlynn
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Freddie Bray
- Section of Cancer Surveillance, International Agency for Research on Cancer, Lyon, France
| |
Collapse
|
28
|
Bastrup FA, Vissing M, Gehl J. Electrochemotherapy with intravenous bleomycin for patients with cutaneous malignancies, across tumour histology: a systematic review. Acta Oncol 2022; 61:1093-1104. [DOI: 10.1080/0284186x.2022.2110385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Freya A. Bastrup
- Department of Clinical Oncology and Palliative Care, Zealand University Hospital, Roskilde, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mille Vissing
- Department of Clinical Oncology and Palliative Care, Zealand University Hospital, Roskilde, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Julie Gehl
- Department of Clinical Oncology and Palliative Care, Zealand University Hospital, Roskilde, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
29
|
Ramanathan S, Prasad M, Vora T, Parambil BC, Kembhavi S, Ramadwar M, Khanna N, Laskar S, Kurkure P, Qureshi S, Banavali S, Chinnaswamy G. Outcomes and prognostic variables of extracranial germ cell tumors in children and adolescents treated over a decade: A developing world perspective. Pediatr Blood Cancer 2022; 69:e29765. [PMID: 35561025 DOI: 10.1002/pbc.29765] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/11/2022] [Accepted: 04/18/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND The purpose of this single-center study was to analyze the outcomes of extracranial germ cell tumors (GCTs) in children treated on a multimodality regimen. METHODS Retrospective study of children (<18 years) with a histopathologically confirmed diagnosis of extracranial GCT over a period of 10 years (January 2009 to December 2018) treated on a uniform institution-based protocol consisting of both cisplatin- and carboplatin-based regimens. All completely excised teratomas and stage I gonadal tumors received no further therapy (low risk [LR]); stage IV ovarian, stage III-IV extragonadal GCTs received six cycles of chemotherapy (high risk [HR]), and the remaining received four cycles of chemotherapy (intermediate risk [IR]). RESULTS A total of 297 children were treated with a female:male ratio of 1.72:1 and median age of 4 years. Forty-three children had pure teratomas. Gonadal GCTs (N = 180) were more common than extragonadal GCTs (N = 117) with ovary as primary site in 128 children (43%) and sacrococcygeal site being the commonest extragonadal location (N = 41; 14%). LR, IR, and HR disease were noted in 60 (20.2%), 125 (42%), and 112 (37.8%) patients, respectively. Three-fourths of ovarian tumors and half of testicular tumors operated prior to presentation needed upstaging. Forty-one patients relapsed and 43 children expired (disease-related: 33; toxic deaths: 9; unknown: 1). The 5-year event-free survival (EFS)/overall survival (OS) of malignant GCT (n = 254) was 72.50%/82.70%, respectively, with gonadal site (p = .001), LR and IR (p = .001) and nonmetastatic disease (p = .001) being favorable prognostic variables. CONCLUSIONS The LR and IR GCTs in our cohort had an excellent outcome. A significant proportion of IR gonadal GCTs can be spared of systemic chemotherapy by adhering to strict surgical guidelines. In HR GCTs however, intensifying therapies to improve outcomes must be balanced against the risk of cumulative toxicity, more so in a resource-limited setting.
Collapse
Affiliation(s)
| | - Maya Prasad
- Department of Pediatric Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Tushar Vora
- Department of Pediatric Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Badira C Parambil
- Department of Pediatric Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Seema Kembhavi
- Department of Radiodiagnosis, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Mukta Ramadwar
- Department of Pathology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Nehal Khanna
- Department of Radiation Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Siddhartha Laskar
- Department of Radiation Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Purna Kurkure
- Department of Pediatric Hematology/Oncology, SRCC Children's Hospital, Mumbai, Maharashtra, India
| | - Sajid Qureshi
- Department of Pediatric Surgery, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Shripad Banavali
- Department of Pediatric Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Girish Chinnaswamy
- Department of Pediatric Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| |
Collapse
|
30
|
Papachristofilou A, Zimmermann F. [Malignant testicular neoplasms in the cisplatin era: causes of death and mortality in a cohort study]. Strahlenther Onkol 2022; 198:862-865. [PMID: 35581422 DOI: 10.1007/s00066-022-01957-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Alexandros Papachristofilou
- Klinik für Strahlentherapie und Radioonkologie, Universitätsspital Basel, Petersgraben 4, 4031, Basel, Schweiz
| | - Frank Zimmermann
- Klinik für Strahlentherapie und Radioonkologie, Universitätsspital Basel, Petersgraben 4, 4031, Basel, Schweiz.
| |
Collapse
|
31
|
Ebenezer O, Shapi M, Tuszynski JA. A Review of the Recent Developments of Molecular Hybrids Targeting Tubulin Polymerization. Int J Mol Sci 2022; 23:4001. [PMID: 35409361 PMCID: PMC8999808 DOI: 10.3390/ijms23074001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/17/2022] [Accepted: 03/29/2022] [Indexed: 12/17/2022] Open
Abstract
Microtubules are cylindrical protein polymers formed from αβ-tubulin heterodimers in the cytoplasm of eukaryotic cells. Microtubule disturbance may cause cell cycle arrest in the G2/M phase, and anomalous mitotic spindles will form. Microtubules are an important target for cancer drug action because of their critical role in mitosis. Several microtubule-targeting agents with vast therapeutic advantages have been developed, but they often lead to multidrug resistance and adverse side effects. Thus, single-target therapy has drawbacks in the effective control of tubulin polymerization. Molecular hybridization, based on the amalgamation of two or more pharmacophores of bioactive conjugates to engender a single molecular structure with enhanced pharmacokinetics and biological activity, compared to their parent molecules, has recently become a promising approach in drug development. The practical application of combined active scaffolds targeting tubulin polymerization inhibitors has been corroborated in the past few years. Meanwhile, different designs and syntheses of novel anti-tubulin hybrids have been broadly studied, illustrated, and detailed in the literature. This review describes various molecular hybrids with their reported structural-activity relationships (SARs) where it is possible in an effort to generate efficacious tubulin polymerization inhibitors. The aim is to create a platform on which new active scaffolds can be modeled for improved tubulin polymerization inhibitory potency and hence, the development of new therapeutic agents against cancer.
Collapse
Affiliation(s)
- Oluwakemi Ebenezer
- Department of Chemistry, Faculty of Natural Science, Mangosuthu University of Technology, Umlazi 4031, South Africa; (O.E.); (M.S.)
| | - Michael Shapi
- Department of Chemistry, Faculty of Natural Science, Mangosuthu University of Technology, Umlazi 4031, South Africa; (O.E.); (M.S.)
| | - Jack A. Tuszynski
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
- Department of Physics, University of Alberta, Edmonton, AB T6G 2E1, Canada
- DIMEAS, Politecnico di Torino, 10129 Turin, Italy
| |
Collapse
|
32
|
Aldrink JH, Glick RD, Baertschiger RM, Kulaylat AN, Lautz TB, Christison-Lagay E, Grant CN, Tracy E, Dasgupta R, Brown EG, Mattei P, Rothstein DH, Rodeberg DA, Ehrlich PF. Update on pediatric testicular germ cell tumors. J Pediatr Surg 2022; 57:690-699. [PMID: 33975708 DOI: 10.1016/j.jpedsurg.2021.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/25/2021] [Accepted: 04/01/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Testicular germ cell tumors are uncommon tumors that are encountered by pediatric surgeons and urologists and require a knowledge of appropriate contemporary evaluation and surgical and medical management. METHOD A review of the recommended diagnostic evaluation and current surgical and medical management of children and adolescents with testicular germ cell tumors based upon recently completed clinical trials was performed and summarized in this article. RESULTS In this summary of childhood and adolescent testicular germ cell tumors, we review the initial clinical evaluation, surgical and medical management, risk stratification, results from recent prospective cooperative group studies, and clinical outcomes. A summary of recently completed clinical trials by pediatric oncology cooperative groups is provided, and best surgical practices are discussed. CONCLUSIONS Testicular germ cell tumors in children are rare tumors. International collaborations, data-sharing, and enrollment of patients at all stages and risk classifications into active clinical trials will enhance our knowledge of these rare tumors and most importantly improve outcomes of patients with testicular germ cell tumors. LEVEL OF EVIDENCE This is a review article of previously published and referenced level 1 and 2 studies, but also includes expert opinion level 5, represented by the American Pediatric Surgical Association Cancer Committee.
Collapse
Affiliation(s)
- Jennifer H Aldrink
- Department of Surgery, Division of Pediatric Surgery, The Ohio State University College of Medicine, Nationwide Children's Hospital, Columbus, OH 43205, United States.
| | - Richard D Glick
- Division of Pediatric Surgery, Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Cohen Children's Medical Center, New Hyde Park, NY, United States
| | - Reto M Baertschiger
- Division of General and Thoracic Surgery, The Hospital for Sick Kids, University of Toronto, Toronto, Ontario, Canada
| | - Afif N Kulaylat
- Division of Pediatric Surgery, Department of Surgery, Penn State Children's Hospital, Hershey, PA, United States
| | - Timothy B Lautz
- Division of Pediatric Surgery, Department of Surgery, Ann and Robert H Lurie Children's Hospital of Chicago, Northwestern University, Chicago, IL, United States
| | - Emily Christison-Lagay
- Department of Surgery, Division of Pediatric Surgery, Yale-New Haven Children's Hospital, Yale School of Medicine, New Haven, CT, United States
| | - Christa N Grant
- Division of Pediatric Surgery, Department of Surgery, Penn State Children's Hospital, Hershey, PA, United States
| | - Elisabeth Tracy
- Department of Surgery, Division of Pediatric Surgery, Duke University Medical Center, Durham, NC, United States
| | - Roshni Dasgupta
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Medical Center, University of Cincinnati, Cincinnati OH, United States
| | - Erin G Brown
- Division of Pediatric Surgery, Department of Surgery, University of California Davis, Sacramento, CA, United States
| | - Peter Mattei
- General, Thoracic and Fetal Surgery, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - David H Rothstein
- Department of Surgery, Seattle Children's Hospital, University of Washington, Seattle, WA, United States
| | - David A Rodeberg
- Department of Surgery, Division of Pediatric Surgery, East Carolina University, Greenville, NC, United States
| | - Peter F Ehrlich
- Department of Surgery, Mott Children's Hospital, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
33
|
Testicular Germ Cell Tumours and Proprotein Convertases. Cancers (Basel) 2022; 14:cancers14071633. [PMID: 35406405 PMCID: PMC8996948 DOI: 10.3390/cancers14071633] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Despite the high survival rate of the most common neoplasia in young Caucasian men: Testicular Germ Cell Tumors (TGCT), the quality of life of these patients is impaired by the multiple long-term side effects of their treatment. The study of molecules that can serve both as diagnostic biomarkers for tumor development and as therapeutic targets seems necessary. Proprotein convertases (PC) are a group of proteases responsible for the maturation of inactive proproteins with very diverse functions, whose alterations in expression have been associated with various diseases, such as other types of cancer and inflammation. The study of the immune tumor microenvironment and the substrates of PCs could contribute to the development of new and necessary immunotherapies to treat this pathology. Abstract Testicular Germ Cell Tumours (TGCT) are widely considered a “curable cancer” due to their exceptionally high survival rate, even if it is reduced by many years after the diagnosis due to metastases and relapses. The most common therapeutic approach to TGCTs has not changed in the last 50 years despite its multiple long-term side effects, and because it is the most common malignancy in young Caucasian men, much research is needed to better the quality of life of the many survivors. Proprotein Convertases (PC) are nine serine proteases responsible for the maturation of inactive proproteins with many diverse functions. Alterations in their expression have been associated with various diseases, including cancer and inflammation. Many of their substrates are adhesion molecules, metalloproteases and proinflammatory molecules, all of which are involved in tumour development. Inhibition of certain convertases has also been shown to slow tumour formation, demonstrating their involvement in this process. Considering the very established link between PCs and inflammation-related malignancies and the recent studies carried out into the immune microenvironment of TGCTs, the study of the involvement of PCs in testicular cancer may open up avenues for being both a biomarker for diagnosis and a therapeutic target.
Collapse
|
34
|
Hendrikson J, Liu Y, Ng WH, Lee JY, Lim AH, Loh JW, Ng CC, Ong WS, Tan JWS, Tan QX, Ng G, Shannon NB, Lim WK, Lim TK, Chua C, Wong JSM, Tan GHC, So JBY, Yeoh KG, Teh BT, Chia CS, Soo KC, Kon OL, Tan IB, Chan JY, Teo MCC, Ong CAJ. Ligand-mediated PAI-1 inhibition in a mouse model of peritoneal carcinomatosis. Cell Rep Med 2022; 3:100526. [PMID: 35243423 PMCID: PMC8861959 DOI: 10.1016/j.xcrm.2022.100526] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 11/22/2021] [Accepted: 01/19/2022] [Indexed: 12/28/2022]
Abstract
Peritoneal carcinomatosis (PC) present a ubiquitous clinical conundrum in all intra-abdominal malignancies. Via functional and transcriptomic experiments of ascites-treated PC cells, we identify STAT3 as a key signaling pathway. Integrative analysis of publicly available databases and correlation with clinical cohorts (n = 7,359) reveal putative clinically significant activating ligands of STAT3 signaling. We further validate a 3-biomarker prognostic panel in ascites independent of clinical covariates in a prospective study (n = 149). Via single-cell sequencing experiments, we uncover that PAI-1, a key component of the prognostic biomarker panel, is largely secreted by fibroblasts and mesothelial cells. Molecular stratification of ascites using PAI-1 levels and STAT3 activation in ascites-treated cells highlight a therapeutic opportunity based on a phenomenon of paracrine addiction. These results are recapitulated in patient-derived ascites-dependent xenografts. Here, we demonstrate therapeutic proof of concept of direct ligand inhibition of a prognostic target within an enclosed biological space.
Collapse
Affiliation(s)
- Josephine Hendrikson
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Ying Liu
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Wai Har Ng
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Jing Yi Lee
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Abner Herbert Lim
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Jui Wan Loh
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Cedric C.Y. Ng
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Whee Sze Ong
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Joey Wee-Shan Tan
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Qiu Xuan Tan
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Gillian Ng
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Nicholas B. Shannon
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Weng Khong Lim
- SingHealth Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore 169609, Singapore
- Cancer and Stem Biology Signature Research Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Tony K.H. Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169856, Singapore
- Pathology Academic Clinical Program, SingHealth Duke-NUS Academic Medical Centre, Singapore 168753, Singapore
| | - Clarinda Chua
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Jolene Si Min Wong
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Grace Hwei Ching Tan
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
| | - Jimmy Bok Yan So
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Division of Surgical Oncology, National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| | - Khay Guan Yeoh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Department of Gastroenterology and Hepatology, National University Hospital, Singapore 119074, Singapore
| | - Bin Tean Teh
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- Institute of Molecular and Cell Biology, A∗STAR Research Entities, Singapore 138673, Singapore
| | - Claramae Shulyn Chia
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Khee Chee Soo
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
| | - Oi Lian Kon
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Iain Beehuat Tan
- Cancer and Stem Biology Signature Research Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
- Laboratory of Applied Cancer Genomics, Genome Institute of Singapore, A∗STAR Research Entities, Singapore 138672, Singapore
| | - Jason Yongsheng Chan
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Melissa Ching Ching Teo
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
| | - Chin-Ann J. Ong
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Institute of Molecular and Cell Biology, A∗STAR Research Entities, Singapore 138673, Singapore
| | - on behalf of the Singapore Peritoneal Oncology Study (SPOS) Group and Singapore Gastric Cancer Consortium (SGCC)
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- SingHealth Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore 169609, Singapore
- Cancer and Stem Biology Signature Research Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169856, Singapore
- Pathology Academic Clinical Program, SingHealth Duke-NUS Academic Medical Centre, Singapore 168753, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Division of Surgical Oncology, National University Cancer Institute, National University Health System, Singapore 119074, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Department of Gastroenterology and Hepatology, National University Hospital, Singapore 119074, Singapore
- Institute of Molecular and Cell Biology, A∗STAR Research Entities, Singapore 138673, Singapore
- Laboratory of Applied Cancer Genomics, Genome Institute of Singapore, A∗STAR Research Entities, Singapore 138672, Singapore
| |
Collapse
|
35
|
King J, Adra N. Current Status of Stem Cell Transplant in Treatment of Testicular Germ Cell Tumors. Curr Oncol Rep 2022; 24:303-310. [PMID: 35113353 DOI: 10.1007/s11912-022-01204-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE OF REVIEW We aim to discuss the history of high-dose chemotherapy with autologous stem-cell transplant in testicular germ cell tumors, prognostic factors to consider prior to transplant, and issues both during and after transplant while also touching on the use of conventional-dose vs. high-dose chemotherapy for initial salvage treatment in patients with relapsed disease. RECENT FINDINGS The advancements in the treatment of testicular cancer have led to the majority of patients even with distant metastases being cured of their malignancy. Despite this, around 20% of patients with metastatic disease will relapse after first-line therapy, and the majority of these patients will go on to need further salvage chemotherapy, either with conventional-dose chemotherapy or high-dose chemotherapy. High-dose chemotherapy with autologous stem-cell transplant is an effective salvage therapy and will still remarkably result in cures for the majority of patients with relapsed disease. While patients receiving it as even third-line salvage therapy may achieve cures, earlier administration likely results in greater efficacy.
Collapse
Affiliation(s)
- Jennifer King
- Division of Hematology & Medical Oncology - Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, 535 Barnhill Dr, RT 459, Indianapolis, IN, 46202, USA
| | - Nabil Adra
- Division of Hematology & Medical Oncology - Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, 535 Barnhill Dr, RT 459, Indianapolis, IN, 46202, USA.
| |
Collapse
|
36
|
Folk WP, Kumari A, Iwasaki T, Cassimere EK, Pyndiah S, Martin E, Homlar K, Sakamuro D. New Synthetic Lethality Re-Sensitizing Platinum-Refractory Cancer Cells to Cisplatin In Vitro: The Rationale to Co-Use PARP and ATM Inhibitors. Int J Mol Sci 2021; 22:ijms222413324. [PMID: 34948122 PMCID: PMC8704450 DOI: 10.3390/ijms222413324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/30/2021] [Accepted: 12/07/2021] [Indexed: 12/31/2022] Open
Abstract
The pro-apoptotic tumor suppressor BIN1 inhibits the activities of the neoplastic transcription factor MYC, poly (ADP-ribose) polymerase-1 (PARP1), and ATM Ser/Thr kinase (ATM) by separate mechanisms. Although BIN1 deficits increase cancer-cell resistance to DNA-damaging chemotherapeutics, such as cisplatin, it is not fully understood when BIN1 deficiency occurs and how it provokes cisplatin resistance. Here, we report that the coordinated actions of MYC, PARP1, and ATM assist cancer cells in acquiring cisplatin resistance by BIN1 deficits. Forced BIN1 depletion compromised cisplatin sensitivity irrespective of Ser15-phosphorylated, pro-apoptotic TP53 tumor suppressor. The BIN1 deficit facilitated ATM to phosphorylate the DNA-damage-response (DDR) effectors, including MDC1. Consequently, another DDR protein, RNF8, bound to ATM-phosphorylated MDC1 and protected MDC1 from caspase-3-dependent proteolytic cleavage to hinder cisplatin sensitivity. Of note, long-term and repeated exposure to cisplatin naturally recapitulated the BIN1 loss and accompanying RNF8-dependent cisplatin resistance. Simultaneously, endogenous MYC was remarkably activated by PARP1, thereby repressing the BIN1 promoter, whereas PARP inhibition abolished the hyperactivated MYC-dependent BIN1 suppression and restored cisplatin sensitivity. Since the BIN1 gene rarely mutates in human cancers, our results suggest that simultaneous inhibition of PARP1 and ATM provokes a new BRCAness-independent synthetic lethal effect and ultimately re-establishes cisplatin sensitivity even in platinum-refractory cancer cells.
Collapse
Affiliation(s)
- Watson P. Folk
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (W.P.F.); (A.K.); (T.I.)
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA; (E.M.); (K.H.)
| | - Alpana Kumari
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (W.P.F.); (A.K.); (T.I.)
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA; (E.M.); (K.H.)
| | - Tetsushi Iwasaki
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (W.P.F.); (A.K.); (T.I.)
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA; (E.M.); (K.H.)
- Division of Signal Pathways, Biosignal Research Center, Kobe University, Kobe 657, Japan
| | - Erica K. Cassimere
- Department of Biology, College of Science, Engineering and Technology, Texas Southern University, Houston, TX 77004, USA;
| | | | - Elizabeth Martin
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA; (E.M.); (K.H.)
- Department of Pathology, Medical College of Georgia, Augusta University Medical Center, Augusta, GA 30912, USA
| | - Kelly Homlar
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA; (E.M.); (K.H.)
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University Medical Center, Augusta, GA 30912, USA
| | - Daitoku Sakamuro
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (W.P.F.); (A.K.); (T.I.)
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA; (E.M.); (K.H.)
- Correspondence: ; Tel.: +1-706-(721)-1018
| |
Collapse
|
37
|
Fung C, Travis LB. Testicular Cancer Survivorship: Looking Back to Move Forward. J Clin Oncol 2021; 39:3531-3534. [PMID: 34591594 PMCID: PMC8577670 DOI: 10.1200/jco.21.01984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/08/2021] [Indexed: 11/20/2022] Open
Affiliation(s)
- Chunkit Fung
- Division of Hematology and Oncology, Department of Medicine, University of Rochester School of Medicine and Dentistry, James P. Wilmot Cancer Institute, Rochester, NY
| | - Lois B. Travis
- Department of Medicine, Indiana University School of Medicine, Department of Epidemiology Fairbanks School of Public Health, Indianapolis, IN
| |
Collapse
|
38
|
Timmerman DM, Eleveld TF, Gillis AJM, Friedrichs CC, Hillenius S, Remmers TL, Sriram S, Looijenga LHJ. The Role of TP53 in Cisplatin Resistance in Mediastinal and Testicular Germ Cell Tumors. Int J Mol Sci 2021; 22:ijms222111774. [PMID: 34769213 PMCID: PMC8583723 DOI: 10.3390/ijms222111774] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 12/31/2022] Open
Abstract
Germ cell tumors (GCTs) are considered to be highly curable; however, there are major differences in the outcomes related to histology and anatomical localization. GCTs originating from the testis are, overall, sensitive to platinum-based chemotherapy, whereas GCTs originating from the mediastinum show a worse response, which remains largely unexplained. Here, we address the differences among GCTs from two different anatomical locations (testicular versus mediastinal/extragonadal), with a specific focus on the role of the P53 pathway. It was recently shown that GCTs with TP53 mutations most often localize to the mediastinum. To elucidate the underlying mechanism, TP53 knock-out lines were generated in cisplatin-sensitive and -resistant clones of the representative 2102Ep cell line (wild-type TP53 testicular GCT) and NCCIT cell line (hemizygously mutated TP53, mutant TP53 mediastinal GCT). The full knock-out of TP53 in 2102Ep and resistant NCCIT resulted in an increase in cisplatin resistance, suggesting a contributing role for P53, even in NCCIT, in which P53 had been reported to be non-functional. In conclusion, these results suggest that TP53 mutations contribute to the cisplatin-resistant phenotype of mediastinal GCTs and, therefore, are a potential candidate for targeted treatment. This knowledge provides a novel model system to elucidate the underlying mechanism of clinical behavior and possible alternative treatment of the TP53 mutant and mediastinal GCTs.
Collapse
|
39
|
Wen L, Li J, Lai M, Zhou Z, Hu Q, Deng G, Shan C, Ai R, Li H, Lu M, Zhang L, Wu T, Zhu D, Chen Y, Chen L, Cai L, Zhou C. Survival outcomes, hematologic complications and growth impairment after sequential chemoradiotherapy in intracranial NGGCTs: a retrospective study. Strahlenther Onkol 2021; 198:458-467. [PMID: 34673990 DOI: 10.1007/s00066-021-01857-3] [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: 04/06/2021] [Accepted: 09/19/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE This study aimed to evaluate the clinical features, prognostic factors, and survival outcomes for patients with intracranial nongerminomatous germ cell tumors (NGGCTs), with a particular focus on treatment toxicity for long-term survivors. METHODS Intracranial NGGCTs treated with platinum-based chemotherapy and craniospinal irradiation (CSI) in our institution were retrospectively analyzed. Hematological complications following sequential chemoradiotherapy as well as height and weight in childhood survivors were evaluated. Plasma growth hormone (GH) concentrations prior to and after radiotherapy were obtained for the comparisons. RESULTS A total of 111 intracranial NGGCTs were included. The 3‑year overall survival (OS) and event-free survival (EFS) rates were 83.5% ± 3.9% and 71.0% ± 4.8%, respectively. A combined treatment modality consisting of ≥ 4 cycles of platinum-based chemotherapy and CSI was associated with an improved OS (P = 0.003) and EFS (P < 0.001). Thrombocytopenia of any grade occurred in 35.4% (34/96) of patients, and the threshold age for an increased risk of thrombocytopenia was 14 years (area under the curve AUC = 0.752, P < 0.0001) as derived from receiver operating characteristic (ROC) analysis. Growth impediment was found in 8 of 56 (14%) patients. The age for receiving radiotherapy was found to inversely correlate with height development, revealing a cut-off age of 11.5 years for risking growth impairment (AUC = 0.806, P = 0.004). Consistently, a significant decline in plasma growth hormone after radiotherapy was observed in patients ≤ 11.5 years (P < 0.01) but not patients > 11.5 years. (P > 0.05). CONCLUSION Our study suggested that a combined treatment modality with at least four cycles of chemotherapy and CSI was safe and effective for patients with intracranial NGGCTs. Radiotherapy should be used with caution for patients < 11.5 years due to growth impairment.
Collapse
Affiliation(s)
- Lei Wen
- Department of Oncology, Guangdong Sanjiu Brain Hospital, 510510, Guangzhou, China
| | - Juan Li
- Department of Oncology, Guangdong Sanjiu Brain Hospital, 510510, Guangzhou, China
| | - Mingyao Lai
- Department of Oncology, Guangdong Sanjiu Brain Hospital, 510510, Guangzhou, China
| | - Zhaoming Zhou
- Department of Oncology, Guangdong Sanjiu Brain Hospital, 510510, Guangzhou, China.,Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou, China
| | - Qingjun Hu
- Department of Oncology, Guangdong Sanjiu Brain Hospital, 510510, Guangzhou, China
| | - Guanhua Deng
- Department of Oncology, Guangdong Sanjiu Brain Hospital, 510510, Guangzhou, China
| | - Changguo Shan
- Department of Oncology, Guangdong Sanjiu Brain Hospital, 510510, Guangzhou, China
| | - Ruyu Ai
- Department of Oncology, Guangdong Sanjiu Brain Hospital, 510510, Guangzhou, China
| | - Hainan Li
- Department of Pathology, Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Ming Lu
- Department of Neurosurgery, Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Liang Zhang
- Department of Neurosurgery, Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Taihua Wu
- Department of Neurosurgery, Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Dan Zhu
- Department of Neurosurgery, Guangdong Sanjiu Brain Hospital, Guangzhou, China
| | - Yuanyuan Chen
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Longhua Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
| | - Linbo Cai
- Department of Oncology, Guangdong Sanjiu Brain Hospital, 510510, Guangzhou, China.
| | - Cheng Zhou
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China.
| |
Collapse
|
40
|
Meng X, Ahmed M, Courtney KD, Arafat W, Ibrahim I, Margulis V, Nichols C, Bagrodia A. Prophylaxis Against Thromboembolic Events During Chemotherapy for Germ Cell Cancer. Front Oncol 2021; 11:724682. [PMID: 34692501 PMCID: PMC8529113 DOI: 10.3389/fonc.2021.724682] [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: 06/14/2021] [Accepted: 09/22/2021] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Patients with advanced germ cell tumors (GCT) receiving cisplatin-based chemotherapy have high rates of thromboembolic events (TEE) which can negatively affect their overall survival. While primary TEE prophylaxis during chemotherapy may prevent these events, it is unclear which patients will benefit in this setting. MATERIALS AND METHODS A review of PubMed/Medline was conducted in December 2020 and all pertinent articles were evaluated for relevancy and quality of data for inclusion in the review. RESULTS Studies on patients receiving initial cisplatin-based chemotherapy for advanced GCT have reported up to a 19% rate of TEE. This high rate may be associated with multiple factors including retroperitoneal lymphadenopathy, advanced clinical stage, high risk Khorana scores and presence of a central line. Large phase III clinical trials have demonstrated the benefit of low-molecular-weight-heparin and direct oral anticoagulants for primary prophylaxis and against recurrent TEE. However, primary prophylaxis is currently underutilized with GCT patients starting chemotherapy. CONCLUSION Precise models to predict TEE risk and consideration of anticoagulation are difficult to develop owing to the relatively uncommon nature of GCT and lack of representation in primary TEE prophylaxis clinical trials. Despite these limitations, we believe that the benefits of prophylactic anticoagulation outweigh the risk of major bleeding in select GCT patients with higher risk of TEE. We have developed a simple algorithm to help guide TEE prophylaxis selection based on patient factors and route of chemotherapy administration. Given the high rate of TEE in GCT patients, we believe better utilization of primary prophylaxis in patient starting cisplatin-based chemotherapy will have clinical benefit.
Collapse
Affiliation(s)
- Xiaosong Meng
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Murtaza Ahmed
- School of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Kevin D Courtney
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Waddah Arafat
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Ibrahim Ibrahim
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Vitaly Margulis
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Craig Nichols
- Testicular Cancer Commons, Portland, OR, United States
| | - Aditya Bagrodia
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Urology, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
41
|
Abstract
Testicular germ cell tumours (TGCTs) are the most common solid tumours in young men and have an excellent overall cure rate and prognosis. In most patients, localised disease is cured by surgery alone, and a minority of patients receive short-course adjuvant chemotherapy to reduce the risk of further relapse. Also, in about 80% of patients, metastatic disease can be cured by systemic cisplatin-based chemotherapy. Unfortunately, for a proportion of patients, the disease exhibits platinum resistance and relapse occurs. Despite further lines of systemic treatment, cure can be difficult to achieve in these patients and ultimately about 20% of them will die from disease progression. Addressing the mechanisms underpinning platinum resistance is critical to improving the survival and chances of cure for these patients. This review describes the latest advances in TGCT research, focusing on the identification of novel biomarkers, genetic characteristics and exploring novel treatments.
Collapse
Affiliation(s)
- Teresa Mele
- The Royal Marsden NHS Foundation Trust, Sutton, UK
- The Institute of Cancer Research, London, UK
| | - Alison Reid
- The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Robert Huddart
- The Royal Marsden NHS Foundation Trust, Sutton, UK
- The Institute of Cancer Research, London, UK
| |
Collapse
|
42
|
Mapuskar KA, Steinbach EJ, Zaher A, Riley DP, Beardsley RA, Keene JL, Holmlund JT, Anderson CM, Zepeda-Orozco D, Buatti JM, Spitz DR, Allen BG. Mitochondrial Superoxide Dismutase in Cisplatin-Induced Kidney Injury. Antioxidants (Basel) 2021; 10:antiox10091329. [PMID: 34572961 PMCID: PMC8469643 DOI: 10.3390/antiox10091329] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023] Open
Abstract
Cisplatin is a chemotherapy agent commonly used to treat a wide variety of cancers. Despite the potential for both severe acute and chronic side effects, it remains a preferred therapeutic option for many malignancies due to its potent anti-tumor activity. Common cisplatin-associated side-effects include acute kidney injury (AKI) and chronic kidney disease (CKD). These renal injuries may cause delays and potentially cessation of cisplatin therapy and have long-term effects on renal function reserve. Thus, developing mechanism-based interventional strategies that minimize cisplatin-associated kidney injury without reducing efficacy would be of great benefit. In addition to its action of cross-linking DNA, cisplatin has been shown to affect mitochondrial metabolism, resulting in mitochondrially derived reactive oxygen species (ROS). Increased ROS formation in renal proximal convoluted tubule cells is associated with cisplatin-induced AKI and CKD. We review the mechanisms by which cisplatin may induce AKI and CKD and discuss the potential of mitochondrial superoxide dismutase mimetics to prevent platinum-associated nephrotoxicity.
Collapse
Affiliation(s)
- Kranti A. Mapuskar
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Emily J. Steinbach
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Amira Zaher
- Biomedical Science Program, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA;
| | - Dennis P. Riley
- Galera Therapeutics, Inc., Malvern, PA 19355, USA; (D.P.R.); (R.A.B.); (J.L.K.); (J.T.H.)
| | - Robert A. Beardsley
- Galera Therapeutics, Inc., Malvern, PA 19355, USA; (D.P.R.); (R.A.B.); (J.L.K.); (J.T.H.)
| | - Jeffery L. Keene
- Galera Therapeutics, Inc., Malvern, PA 19355, USA; (D.P.R.); (R.A.B.); (J.L.K.); (J.T.H.)
| | - Jon T. Holmlund
- Galera Therapeutics, Inc., Malvern, PA 19355, USA; (D.P.R.); (R.A.B.); (J.L.K.); (J.T.H.)
| | - Carryn M. Anderson
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Diana Zepeda-Orozco
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
- College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Nephrology, Department of Pediatrics, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - John M. Buatti
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Bryan G. Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
- Correspondence: ; Tel.: +1-319-335-8019; Fax: +1-319-335-8039
| |
Collapse
|
43
|
Hellesnes R, Myklebust TÅ, Fosså SD, Bremnes RM, Karlsdottir Á, Kvammen Ø, Tandstad T, Wilsgaard T, Negaard HFS, Haugnes HS. Testicular Cancer in the Cisplatin Era: Causes of Death and Mortality Rates in a Population-Based Cohort. J Clin Oncol 2021; 39:3561-3573. [PMID: 34388002 DOI: 10.1200/jco.21.00637] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Using complete information regarding testicular cancer (TC) treatment burden, this study aimed to investigate cause-specific non-TC mortality with impact on previous treatment with platinum-based chemotherapy (PBCT) or radiotherapy (RT). METHODS Overall, 5,707 men identified by the Cancer Registry of Norway diagnosed with TC from 1980 to 2009 were included in this population-based cohort study. By linking data with the Norwegian Cause of Death Registry, standardized mortality ratios (SMRs), absolute excess risks (AERs; [(observed number of deaths - expected number of deaths)/person-years of observation] ×10,000), and adjusted hazard ratios (HRs) were calculated. RESULTS Median follow-up was 18.7 years, during which non-TC death was registered for 665 (12%) men. Overall excess non-TC mortality was 23% (SMR, 1.23; 95% CI, 1.14 to 1.33; AER, 11.14) compared with the general population, with increased risks after PBCT (SMR, 1.23; 95% CI, 1.07 to 1.43; AER, 7.68) and RT (SMR, 1.28; 95% CI, 1.15 to 1.43; AER, 19.55). The highest non-TC mortality was observed in those < 20 years at TC diagnosis (SMR, 2.27; 95% CI, 1.32 to 3.90; AER, 14.42). The most important cause of death was non-TC second cancer with an overall SMR of 1.53 (95% CI, 1.35 to 1.73; AER, 7.94), with increased risks after PBCT and RT. Overall noncancer mortality was increased by 15% (SMR, 1.15; 95% CI, 1.04 to 1.27; AER, 4.71). Excess suicides appeared after PBCT (SMR, 1.65; 95% CI, 1.01 to 2.69; AER, 1.39). Compared with surgery, increased non-TC mortality appeared after 3 (HR, 1.47; 95% CI, 0.91 to 2.39), 4 (HR, 1.41; 95% CI, 1.01 to 1.99), and more than four (HR, 2.04; 95% CI, 1.25 to 3.35) cisplatin-based chemotherapy cycles after > 10 years of follow-up. CONCLUSION TC treatment with PBCT or RT is associated with a significant excess risk of non-TC mortality, and increased risks emerged after more than two cisplatin-based chemotherapy cycles after > 10 years of follow-up.
Collapse
Affiliation(s)
- Ragnhild Hellesnes
- Department of Oncology, University Hospital of North Norway, Tromsø, Norway.,Department of Clinical Medicine, UiT The Arctic University, Tromsø, Norway
| | - Tor Åge Myklebust
- Department of Research and Innovation, Møre and Romsdal Hospital Trust, Ålesund, Norway.,Department of Registration, Cancer Registry of Norway, Oslo, Norway
| | - Sophie D Fosså
- Department of Registration, Cancer Registry of Norway, Oslo, Norway.,Department of Oncology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Roy M Bremnes
- Department of Oncology, University Hospital of North Norway, Tromsø, Norway.,Department of Clinical Medicine, UiT The Arctic University, Tromsø, Norway
| | - Ása Karlsdottir
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Øivind Kvammen
- Department of Oncology, Ålesund Hospital, Ålesund, Norway
| | - Torgrim Tandstad
- The Cancer Clinic, St Olavs University Hospital, Trondheim, Norway.,Department of Clinical and Molecular Medicine, The Norwegian University of Science and Technology, Trondheim, Norway
| | - Tom Wilsgaard
- Department of Community Medicine, UiT The Arctic University, Tromsø, Norway
| | | | - Hege S Haugnes
- Department of Oncology, University Hospital of North Norway, Tromsø, Norway.,Department of Clinical Medicine, UiT The Arctic University, Tromsø, Norway
| |
Collapse
|
44
|
King J, Adra N, Einhorn LH. Testicular cancer: Biology to bedside. Cancer Res 2021; 81:5369-5376. [PMID: 34380632 DOI: 10.1158/0008-5472.can-21-1452] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/27/2021] [Accepted: 08/10/2021] [Indexed: 11/16/2022]
Abstract
Testicular cancer is the first solid tumor with a remarkably high cure rate. This success was only made possible through collaborative efforts of basic and clinical research. Most patients with distant metastases can be cured. However, the majority of these patients are diagnosed at a young age, leaving many decades for the development of treatment-related complications. This has magnified the importance of research into survivorship issues after exposure to platinum-based chemotherapy. This research, along with research into newer biomarkers that will aid in the diagnosis and surveillance of patients and survivors of testicular cancer, will continue to advance the field and provide new opportunities for these patients. There also remains the need for further therapeutic options for patients who unfortunately do not respond to standard treatment regimens and ultimately die from this disease, including a cohort of patients with late relapses and platinum-refractory disease. Here we discuss the advancements in management that led to a highly curable malignancy, while highlighting difficult situations still left to solve as well as emerging research into novel biomarkers.
Collapse
Affiliation(s)
- Jennifer King
- Hematology/Oncology, Indiana University School of Medicine
| | - Nabil Adra
- Hematology/Oncology, Indiana University School of Medicine
| | | |
Collapse
|
45
|
Agrawal V, Abonour R, Abu Zaid M, Althouse SK, Ashkar R, Albany C, Hanna NH, Einhorn LH, Adra N. Survival outcomes and toxicity in patients 40 years old or older with relapsed metastatic germ cell tumors treated with high-dose chemotherapy and peripheral blood stem cell transplantation. Cancer 2021; 127:3751-3760. [PMID: 34260067 DOI: 10.1002/cncr.33771] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND High-dose chemotherapy (HDCT) plus peripheral blood stem cell transplantation (PBSCT) is effective salvage therapy for relapsed metastatic germ cell tumors (GCTs) but has potential toxicity. Historically, an age of ≥40 years has been associated with greater toxicity and worse outcomes. METHODS This is a retrospective analysis of 445 consecutive patients with relapsed GCT treated with HDCT and PBSCT with tandem cycles at Indiana University from between 2004-2017 per our institutional regimen. Kaplan-Meier methods and log-rank tests were used for progression-free survival (PFS) and overall survival (OS) analysis. RESULTS A total of 329 patients were <40 years of age, whereas 116 patients were ≥40 years of age; HDCT was used as second-line therapy in 85% and 79%, respectively. Median follow-up time was 42.5 months (range, 0.3-173.4 months). Grade ≥3 toxicities were similar between either group, except for greater pulmonary (P = .02) and renal toxicity (P = .01) in the ≥40-years-of-age group. Treatment-related mortality was similar between both age groups: 10 patients (3%) in the <40-years-of-age group and 4 patients (3.5%) in ≥40-years-of-age group died from complications of HDCT. Two-year PFS for <40 years of age versus ≥40 years of age was 58.7% versus 59.6% (P = .76) and 2-year OS was 63.9% versus 61.5% (P = .93). Factors predicting worse PFS included Eastern Cooperative Oncology Group performance status ≥1, platinum refractory disease, nonseminoma histology, and not completing 2 cycles of HDCT. Age was not an independent predictor of worse outcomes. CONCLUSIONS HDCT plus PBSCT is effective salvage therapy in patients ≥40 years of age with relapsed metastatic GCT. Patients ≥40 years of age experience similar rates of toxicity and treatment-related mortality as those <40 years of age.
Collapse
Affiliation(s)
- Vaibhav Agrawal
- Division of Hematology-Oncology, Indiana University Simon Comprehensive Cancer Center, Indianapolis, Indiana
| | - Rafat Abonour
- Division of Hematology-Oncology, Indiana University Simon Comprehensive Cancer Center, Indianapolis, Indiana
| | - Mohammad Abu Zaid
- Division of Hematology-Oncology, Indiana University Simon Comprehensive Cancer Center, Indianapolis, Indiana
| | - Sandra K Althouse
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, Indiana
| | - Ryan Ashkar
- Division of Hematology-Oncology, Indiana University Simon Comprehensive Cancer Center, Indianapolis, Indiana
| | - Costantine Albany
- Division of Hematology-Oncology, Indiana University Simon Comprehensive Cancer Center, Indianapolis, Indiana
| | - Nasser H Hanna
- Division of Hematology-Oncology, Indiana University Simon Comprehensive Cancer Center, Indianapolis, Indiana
| | - Lawrence H Einhorn
- Division of Hematology-Oncology, Indiana University Simon Comprehensive Cancer Center, Indianapolis, Indiana
| | - Nabil Adra
- Division of Hematology-Oncology, Indiana University Simon Comprehensive Cancer Center, Indianapolis, Indiana
| |
Collapse
|
46
|
Retinal toxicities of systemic anticancer drugs. Surv Ophthalmol 2021; 67:97-148. [PMID: 34048859 DOI: 10.1016/j.survophthal.2021.05.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 05/08/2021] [Accepted: 05/10/2021] [Indexed: 01/07/2023]
Abstract
Newer anticancer drugs have revolutionized cancer treatment in the last decade, but conventional chemotherapy still occupies a central position in many cancers, with combination therapy and newer methods of delivery increasing their efficacy while minimizing toxicities. We discuss the retinal toxicities of anticancer drugs with an emphasis on the mechanism of toxicity. Uveitis is seen with the use of v-raf murine sarcoma viral oncogene homolog B editing anticancer inhibitors as well as immunotherapy. Most of the cases are mild with only anterior uveitis, but severe cases of posterior uveitis, panuveitis, and Vogt-Koyanagi-Harada-like disease may also occur. In the retina, a transient neurosensory detachment is observed in almost all patients on mitogen-activated protein kinase kinase (MEK) inhibitors. Microvasculopathy is often seen with interferon α, but vascular occlusion is a more serious toxicity caused by interferon α and MEK inhibitors. Crystalline retinopathy with or without macular edema may occur with tamoxifen; however, even asymptomatic patients may develop cavitatory spaces seen on optical coherence tomography. A unique macular edema with angiographic silence is characteristic of taxanes. Delayed dark adaptation has been observed with fenretinide. Interestingly, this drug is finding potential application in Stargardt disease and age-related macular degeneration.
Collapse
|
47
|
Hajdu SI. Pathfinders in oncology from the time the causal relation between tobacco use and lung cancer was established to publication of the first Cancer Staging Manual by the American Joint Committee on Cancer. Cancer 2021; 127:2828-2854. [PMID: 33970484 DOI: 10.1002/cncr.33561] [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: 01/25/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 11/06/2022]
Abstract
During the period from 1962 to 1977, several antigens, notably carcinoembryonic antigen and prostate-specific antigen, were discovered and entered clinical use. Ultrasonography, positron emission tomography scanning, and magnetic resonance imaging were introduced, and adjuvant radiation and chemotherapy after limited surgery became routine procedures. Radioimmunoassay and immunohistochemistry techniques were standardized. The announcement in England and the United States that tobacco is a potent lung carcinogen was long delayed, important news. The US Cancer Act of 1971 made it possible to experiment with newly discovered drugs, transfer promising therapeutic agents from the laboratory to the clinic, and finance randomized clinical trials. Oncologists achieved a series of successes with combination chemotherapy in childhood cancers, adult lymphomas, and testis tumors. Clinical trials demonstrated that breast-conserving therapy is as effective as mastectomy. The discovery of retroviruses, reverse transcriptase, and vascular endothelial growth factor was coupled with learning about oncogenes. The 2-hit theory and the reciprocal translocation of chromosomes helped to solve some of the riddles of oncogenesis. The staging classification of cancers by the American Joint Committee on Cancer unified clinical and pathologic handling and prognostication of malignant tumors. The progress made in oncology between 1962 and 1977 came about through the dedicated work of many individuals. However, there were 9 pathfinders (3 medical oncologists, 2 surgeons, 1 medical nuclear physicist, 1 pediatrician geneticist, 1 hematologist geneticist, and 1 virologist) who, despite their diverse backgrounds, personalities, and interest, made extraordinary contributions to oncology.
Collapse
|
48
|
Masterson TA, Tagawa ST. A 25-year review of advances in testicular cancer: Perspectives on evaluation, treatment, and future directions/challenges. Urol Oncol 2021; 39:561-568. [PMID: 33853746 DOI: 10.1016/j.urolonc.2021.02.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 10/21/2022]
Abstract
The year 2020 will be remembered for a number of different events, both good and bad. For the journal Urologic Oncology, Seminars and Original Investigations, this year represents the 25th anniversary of its inception and 1st publication. Under the encouragement of Editor-in-Chief Dr. Michael Droller, the collective editorial board has put together a reflection of the progresses made among the spectrum of genitourinary cancers across the entirety of therapeutic disciplines. In this review, we discuss the advances achieved in our knowledge and understanding of testicular germ cell tumors since 1995, and the challenges that lie ahead.
Collapse
Affiliation(s)
- Timothy A Masterson
- Department of Urology, Indiana University School of Medicine, Indianapolis, IN.
| | - Scott T Tagawa
- Division of Medical Oncology, Department of Medicine and Department of Urology, Weill Cornell University Medicine, New York, NY
| |
Collapse
|
49
|
Chovanec M, Lauritsen J, Bandak M, Oing C, Kier GG, Kreiberg M, Rosenvilde J, Wagner T, Bokemeyer C, Daugaard G. Late adverse effects and quality of life in survivors of testicular germ cell tumour. Nat Rev Urol 2021; 18:227-245. [PMID: 33686290 DOI: 10.1038/s41585-021-00440-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2021] [Indexed: 02/06/2023]
Abstract
Currently, ~95% of patients with testicular germ cell tumour (TGCT) are cured, resulting in an increasing number of TGCT survivors. Although cured, these men face potential late adverse effects and reduced quality of life. Survivors face a twofold increased risk of second malignant neoplasms after chemotherapy and radiotherapy, with evidence of dose-dependent associations. For survivors managed with surveillance or treated with radiotherapy, the risk of cardiovascular disease (CVD) is comparable to the risk in the general population, whereas treatment with chemotherapy increases the risk of life-threatening CVD, especially during treatment and after 10 years of follow-up. Other adverse effects are organ-related toxicities such as neuropathy and ototoxicity. Pulmonary and renal impairment in patients with TGCT treated with chemotherapy is limited. Survivors of TGCT might experience psychosocial distress including anxiety disorders, fear of cancer recurrence and TGCT-specific issues, such as sexual dysfunction. Late adverse effects can be avoided in most patients with stage I disease if followed on a surveillance programme. However, patients with disseminated disease can experience toxicities associated with radiotherapy and chemotherapy, and/or adverse effects related to surgery for residual disease. The severity of adverse effects increases with dose of both chemotherapy and radiotherapy. This Review discusses the most recent data concerning the late adverse effects of today's standard treatments for TGCT.
Collapse
Affiliation(s)
- Michal Chovanec
- 2nd Department of Oncology, Comenius University, National Cancer Institute, Bratislava, Slovakia
| | - Jakob Lauritsen
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Mikkel Bandak
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Christoph Oing
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gry Gundgaard Kier
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Michael Kreiberg
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Josephine Rosenvilde
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Thomas Wagner
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gedske Daugaard
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
50
|
McSweeney KR, Gadanec LK, Qaradakhi T, Ali BA, Zulli A, Apostolopoulos V. Mechanisms of Cisplatin-Induced Acute Kidney Injury: Pathological Mechanisms, Pharmacological Interventions, and Genetic Mitigations. Cancers (Basel) 2021; 13:1572. [PMID: 33805488 PMCID: PMC8036620 DOI: 10.3390/cancers13071572] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023] Open
Abstract
Administration of the chemotherapeutic agent cisplatin leads to acute kidney injury (AKI). Cisplatin-induced AKI (CIAKI) has a complex pathophysiological map, which has been linked to cellular uptake and efflux, apoptosis, vascular injury, oxidative and endoplasmic reticulum stress, and inflammation. Despite research efforts, pharmaceutical interventions, and clinical trials spanning over several decades, a consistent and stable pharmacological treatment option to reduce AKI in patients receiving cisplatin remains unavailable. This has been predominately linked to the incomplete understanding of CIAKI pathophysiology and molecular mechanisms involved. Herein, we detail the extensively known pathophysiology of cisplatin-induced nephrotoxicity that manifests and the variety of pharmacological and genetic alteration studies that target them.
Collapse
|