14501
|
Huang J, Guo P, Moses MA. Rationally Designed Antibody Drug Conjugates Targeting the Breast Cancer-Associated Endothelium. ACS Biomater Sci Eng 2019; 6:2563-2569. [PMID: 33463296 DOI: 10.1021/acsbiomaterials.9b01060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The promise of antiangiogenic therapy for the treatment of breast cancer has been limited by the inability to selectively disrupt the established tumor vasculature. Here, we report the development of rationally designed antibody drug conjugates (ADCs) that can selectively recognize and attack breast tumor-associated endothelial cells (BTECs), while sparing normal endothelial cells (NECs). We first performed a quantitative and unbiased screening of a panel of cancer-related antigens on human BTECs and identified CD105 as the optimal ADC target on these cells. We then used clinically approved ADC linkers and cytotoxic drugs to engineer two CD105-targeted ADCs: CD105-DM1 and CD105-MMAE and evaluated their in vitro efficacy in human BTECs and NECs. We found that both CD105-DM1 and CD105-MMAE exhibited highly potent and selective cytotoxicity against BTECs with IC50 values of 3.2 and 3.7 nM, respectively, significantly lower than their IC50 values on NECs (8-13 fold). Our proof-of-principle study suggests that CD105-targeted ADCs are promising antiangiogenic agents that have the potential to be used to inhibit the established tumor vasculature of breast tumors in a safe and precise manner.
Collapse
Affiliation(s)
- Jing Huang
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, United States.,Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Peng Guo
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, United States.,Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Marsha A Moses
- Vascular Biology Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, United States.,Department of Surgery, Harvard Medical School and Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
14502
|
Nadalin S, Königsrainer A, Capobianco I, Settmacher U, Rauchfuss F. Auxiliary living donor liver transplantation combined with two-stage hepatectomy for unresectable colorectal liver metastases. Curr Opin Organ Transplant 2019; 24:651-658. [PMID: 31397729 DOI: 10.1097/mot.0000000000000695] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW To review the role of liver transplantation for unresectable colorectal liver metastases (u-CRLM) and to describe the intial experience with auxiliary living donor liver transplantation combined with two-stage hepatectomy for u-CRLM (i.e. living donor RAPID). RECENT FINDINGS Patients affected with u-CRLM have a poor prognosis with 5 years overall survival (OS) rate less than 10% under standard modern chemotherapy.There is an actual international consensus that liver transplantation for u-CRLM represents a viable option in highly selected patients with OS rate at 5 years up to 80% notwithstanding high recurrence rates. Due to the scarcity of whole liver graft from deceased donors, the RAPID procedure (i.e. resection and partial liver segment 2-3 transplantation from deceased donors with delayed total hepatectomy) has been introduced as possible alternative. The RAPID procedure represents the most actual and modern fusion of the two most challenging procedures of modern hepatobiliary and liver transplant surgery: that is auxiliary partial orthotopic liver transplantation and associating liver partition and portal vein ligation for staged hepatectomy. Although the deceased donor-RAPID procedure may show promising results, the basic problem of scarcity of organs from deceased donors and mainly the lack of splittable organs still remains. SUMMARY The living donor RAPID, based on transplantation of left lateral segments from living donor, may represent the way out to this problem. It is feasible and safe (for both donor and recipient), but characterized by a very challenging high-end transplantological procedure.
Collapse
Affiliation(s)
- Silvio Nadalin
- Department of General, Visceral and Transplant Surgery, University Hospital Tuebingen, Tubingen
| | - Alfred Königsrainer
- Department of General, Visceral and Transplant Surgery, University Hospital Tuebingen, Tubingen
| | - Ivan Capobianco
- Department of General, Visceral and Transplant Surgery, University Hospital Tuebingen, Tubingen
| | - Utz Settmacher
- Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| | - Falk Rauchfuss
- Department of General, Visceral and Vascular Surgery, University Hospital Jena, Jena, Germany
| |
Collapse
|
14503
|
Zhang W, Song S, Wang H, Wang Q, Li D, Zheng S, Xu Z, Zhang H, Wang J, Sun J. In vivo irreversible albumin-binding near-infrared dye conjugate as a naked-eye and fluorescence dual-mode imaging agent for lymph node tumor metastasis diagnosis. Biomaterials 2019; 217:119279. [DOI: 10.1016/j.biomaterials.2019.119279] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/30/2019] [Accepted: 06/12/2019] [Indexed: 01/22/2023]
|
14504
|
Fang Y, Pu N, Zhang L, Wu W, Lou W. Chemoradiotherapy is associated with improved survival for resected pancreatic adenosquamous carcinoma: a retrospective cohort study from the SEER database. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:522. [PMID: 31807504 DOI: 10.21037/atm.2019.10.12] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background The prognosis of pancreatic adenosquamous carcinoma (PASC) after surgery is poor. The purpose of this study was to clarify the prognostic factors of PASC and evaluate the efficacy of combination chemoradiotherapy. Methods The patients' data retrieved from the Surveillance, Epidemiology, and End Results database (SEER) between 2004 and 2015 were stratified and analyzed in this study. The univariate and multivariate analysis were used for overall survival (OS) and cancer-specific survival (CSS). Results T staging, M staging, chemotherapy and radiotherapy is the independent prognostic indicator after PASC resection for both OS and CSS. In the total cohort, 44 patients had both chemo and radiotherapy, with median OS 23 months and CSS 29 months, which was significantly better than neither chemo nor radiotherapy group (68 patients, median OS 8 months and CSS 11 months), and either chemotherapy or radiotherapy group (91 patients, median OS 13 months and CSS 15 months). The survival benefit of chemoradiotherapy was validated in the specific group (n=159) who had only primary PASC. PASC patients receiving chemoradiotherapy had longer OS and CSS than those with neither chemo nor radiotherapy in TNM stage I, II and IV subgroups. Conclusions The chemoradiotherapy revealed its prognostic superiority in PASC treatment.
Collapse
Affiliation(s)
- Yuan Fang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ning Pu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lei Zhang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wenchuan Wu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wenhui Lou
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
14505
|
Exercise training, circulating cytokine levels and immune function in cancer survivors: A meta-analysis. Brain Behav Immun 2019; 81:92-104. [PMID: 31454519 DOI: 10.1016/j.bbi.2019.08.187] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/06/2019] [Accepted: 08/21/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Anti-cancer therapies lead to chronic non-resolving inflammation and reduced immune function. One potential therapy is exercise training, but the effectiveness of these interventions to improve immune-related outcomes, the gaps in the literature, and recommendations to progress the field need to be determined. OBJECTIVES (1) to conduct separate meta-analyses in cancer survivors to determine the effects of exercise training on pro- and anti-inflammatory markers, and immune cell proportions and function; and (2) to perform subgroup analyses to determine whether exercise modality, cancer type, and specific markers help to explain heterogeneity in each meta-analysis. DATA SOURCES Electronic databases (PubMed/MEDLINE, EMBASE, CENTRAL, and CINAHL) from inception to March 2018. The reference lists of eligible articles and relevant reviews were also checked. STUDY SELECTION Inclusion criteria were adult cancer survivors from randomized controlled trials performing structured exercise intervention (aerobic, resistance or combined training or Tai Chi/yoga) compared to usual care control group and included pro-inflammatory, anti-inflammatory, and/or immune cell outcomes. APPRAISAL AND SYNTHESIS METHODS A total of 5349 potentially eligible articles were identified, of which 26 articles (27 trials) met the inclusion criteria. Effect sizes were calculated as standardized mean differences (SMD), where <0.2 was defined as trivial, 0.2-0.3 as small, 0.4-0.8 as moderate, and >0.8 as a large effect. RESULTS Exercise training decreased pro-inflammatory markers (SMD: -0.2, 95% CI: -0.4, -0.1, p < 0.001). Sub-group analysis for the pro-inflammatory markers indicated that combined aerobic and resistance training had the greatest effect (SMD: -0.3, 95% CI: -0.5, -1.9, p < 0.001), that prostate (SMD: -0.5, 95% CI: -0.8, 0.1, p = 0.004) and breast cancer populations were most responsive (SMD: -0.2, 95% CI: -0.3, -0.1, p = 0.001), and that C-reactive protein (SMD: -0.5, 95% CI: -0.9, -0.06, p = 0.025) and tumor necrosis factor (SMD: -0.3, 95% CI: -0.5, -0.06, p = 0.004) were the most sensitive to change. Exercise training tended to decrease anti-inflammatory markers (p = 0.072) but had no effect on natural killer or natural killer T cell proportions or cytotoxic activity. CONCLUSIONS Exercise training reduces pro-inflammatory markers in cancer survivors, with the strongest evidence for combined training and for prostate and breast cancer survivors. Further research is warranted to determine if these changes are clinically relevant or are associated with improvements in symptoms. To strengthen future research, focusing on novel immune populations that include functional parameters and standardized reporting of key immune outcomes is recommended.
Collapse
|
14506
|
Ganesh K, Wu C, O'Rourke KP, Szeglin BC, Zheng Y, Sauvé CEG, Adileh M, Wasserman I, Marco MR, Kim AS, Shady M, Sanchez-Vega F, Karthaus WR, Won HH, Choi SH, Pelossof R, Barlas A, Ntiamoah P, Pappou E, Elghouayel A, Strong JS, Chen CT, Harris JW, Weiser MR, Nash GM, Guillem JG, Wei IH, Kolesnick RN, Veeraraghavan H, Ortiz EJ, Petkovska I, Cercek A, Manova-Todorova KO, Saltz LB, Lavery JA, DeMatteo RP, Massagué J, Paty PB, Yaeger R, Chen X, Patil S, Clevers H, Berger MF, Lowe SW, Shia J, Romesser PB, Dow LE, Garcia-Aguilar J, Sawyers CL, Smith JJ. A rectal cancer organoid platform to study individual responses to chemoradiation. Nat Med 2019; 25:1607-1614. [PMID: 31591597 PMCID: PMC7385919 DOI: 10.1038/s41591-019-0584-2] [Citation(s) in RCA: 350] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 08/15/2019] [Indexed: 12/22/2022]
Abstract
Rectal cancer (RC) is a challenging disease to treat that requires chemotherapy, radiation and surgery to optimize outcomes for individual patients. No accurate model of RC exists to answer fundamental research questions relevant to patients. We established a biorepository of 65 patient-derived RC organoid cultures (tumoroids) from patients with primary, metastatic or recurrent disease. RC tumoroids retained molecular features of the tumors from which they were derived, and their ex vivo responses to clinically relevant chemotherapy and radiation treatment correlated with the clinical responses noted in individual patients' tumors. Upon engraftment into murine rectal mucosa, human RC tumoroids gave rise to invasive RC followed by metastasis to lung and liver. Importantly, engrafted tumors displayed the heterogenous sensitivity to chemotherapy observed clinically. Thus, the biology and drug sensitivity of RC clinical isolates can be efficiently interrogated using an organoid-based, ex vivo platform coupled with in vivo endoluminal propagation in animals.
Collapse
Affiliation(s)
- Karuna Ganesh
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chao Wu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kevin P O'Rourke
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medicine/Rockefeller University/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Bryan C Szeglin
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Albert Einstein College of Medicine, Bronx, NY, USA
| | - Youyun Zheng
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Mohammad Adileh
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Isaac Wasserman
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael R Marco
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amanda S Kim
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maha Shady
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Francisco Sanchez-Vega
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Computational Oncology Service, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wouter R Karthaus
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Helen H Won
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Seo-Hyun Choi
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Raphael Pelossof
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Afsar Barlas
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Peter Ntiamoah
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Emmanouil Pappou
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Arthur Elghouayel
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James S Strong
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chin-Tung Chen
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jennifer W Harris
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martin R Weiser
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Garrett M Nash
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jose G Guillem
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Iris H Wei
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard N Kolesnick
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Harini Veeraraghavan
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eduardo J Ortiz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Iva Petkovska
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrea Cercek
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Leonard B Saltz
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jessica A Lavery
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ronald P DeMatteo
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Joan Massagué
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Philip B Paty
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rona Yaeger
- Gastrointestinal Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xi Chen
- Department of Public Health Sciences, Sylvestor Comprehensive Cancer Center, Miami, FL, USA
| | - Sujata Patil
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, University of Medical Center, Utrecht, The Netherlands
| | - Michael F Berger
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Scott W Lowe
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jinru Shia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Gastrointestinal Pathology, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul B Romesser
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lukas E Dow
- Sandra and Edward Meyer Cancer Center, Departments of Medicine and Biochemistry, Weill Cornell Medicine, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Julio Garcia-Aguilar
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charles L Sawyers
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - J Joshua Smith
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
14507
|
Yan Q, Zheng WJ, Chen QL, Wang BQ, Luo HY, Xue J, Wang XW. Nomogram to predict overall survival and disease-specific survival with appendiceal mucinous adenocarcinoma. Medicine (Baltimore) 2019; 98:e17332. [PMID: 31577727 PMCID: PMC6783251 DOI: 10.1097/md.0000000000017332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
To predict the survival of appendiceal mucinous adenocarcinoma (AMA) by prognostic nomogram.A total of 3234 patients with AMA were collected from the Surveillance, Epidemiology, and End Results (SEER) database from 1973 to 2015. Univariate and multivariate Cox proportional hazards (PH) regression analyses were used to generate independent prognostic factors. These variables were included in the nomogram to predict overall survival (OS) and disease-specific survival (DSS) at 1-, 3-, and 5- years. These data are validated both internally and externally. The consistency index (C-index) and calibration chart were used to estimate the accuracy of the nomogram.The study cohort was randomly divided into the training (n = 2155) and validation group (n = 1799). According to univariate and multivariate analyses, age at diagnosis, marital status, sex, histological differentiation, SEER extent of disease, number of local lymph nodes examined, whether they were positive, and surgical methods were independent prognostic factors for OS and DSS. These factors were incorporated into the nomogram. Internal validation in the training cohort showed that the C-index values for nomogram predictions of OS and DSS were 0.73 (95% CI 0.70-0.76) and 0.77 (95% CI 0.73-0.81), respectively. Similarly, the corresponding C-index values in the external validation cohort were 0.76 (95% CI 0.70-0.81) and 0.75 (95% CI 0.71-0.80). The Calibration plots revealed that the actual survival and nomogram prediction had a good consistency.Build a nomogram in the SEER database to predict OS and DSS in patients with AMA. It can provide accurate and personalised survival prediction for clinicians and patients.
Collapse
Affiliation(s)
- Qian Yan
- Guangzhou University of Chinese Medicine
| | | | | | | | | | - Jiao Xue
- Guangzhou University of Chinese Medicine
| | - Xiong Wen Wang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
14508
|
Brooks GA, Jhatakia S, Tripp A, Landrum MB, Christian TJ, Newes-Adeyi G, Cafardi S, Hassol A, Simon C, Keating NL. Early Findings From the Oncology Care Model Evaluation. J Oncol Pract 2019; 15:e888-e896. [DOI: 10.1200/jop.19.00265] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE: The Oncology Care Model (OCM) is an alternative payment model administered by the Centers for Medicare & Medicaid Services (CMS) that is structured around 6-month chemotherapy treatment episodes. This report describes the CMS-sponsored OCM evaluation and summarizes early evaluation findings. METHODS: The OCM evaluation examines health care spending and use, quality of care, and patient experience during chemotherapy treatment episodes. Because OCM participation is voluntary, the evaluation compares participating physician practices with a propensity-matched group of nonparticipating practices by using a difference-in-differences approach. This report examines 6-month episodes initiated during the first OCM performance period (July 1, 2016, through January 1, 2017). RESULTS: During the first OCM performance period, there was no statistically significant impact of OCM on total episode payments. There were small declines in intensive care unit (ICU) admissions (7 per 1,000 episodes) and emergency department visits (15 per 1,000 episodes); there was no statistically significant impact on hospitalizations or 30-day readmissions. Analyses of care quality and end-of-life care showed statistically significant impacts of OCM on the proportion of patients with inpatient hospitalizations in the last 30 days of life (1.5% absolute decrease) and ICU admissions in the last 30 days of life (2.1% decrease). There was no significant OCM impact on measures of hospice use. CONCLUSION: Early findings from the OCM evaluation demonstrate modest program-related impacts on some acute care services and no change in total episode payments. Early findings may not reflect practice redesign efforts that were phased in after the beginning of OCM.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Nancy L. Keating
- Harvard Medical School, Boston, MA
- Brigham and Women’s Hospital, Boston, MA
| |
Collapse
|
14509
|
Deshmukh SK, Srivastava SK, Poosarla T, Dyess DL, Holliday NP, Singh AP, Singh S. Inflammation, immunosuppressive microenvironment and breast cancer: opportunities for cancer prevention and therapy. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:593. [PMID: 31807574 DOI: 10.21037/atm.2019.09.68] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Breast cancer is the most commonly diagnosed malignancy and a leading cause of cancer-related death in women worldwide. It also exhibits pronounced racial disparities in terms of incidence and clinical outcomes. There has been a growing interest in research community to better understand the role of the microenvironment in cancer. Several lines of evidence have highlighted the significance of chronic inflammation at the local and/or systemic level in breast tumor pathobiology. Inflammation can influence breast cancer progression, metastasis and therapeutic outcome by establishing a tumor supportive immune microenvironment. These processes are mediated through a variety of cytokines and hormones that exert their biological actions either locally or distantly via systemic circulation. Targeting of immune and inflammatory pathways has met tremendous success in some cancers underscoring the importance of research to further our understanding of these systems in breast cancer. This knowledge can be helpful not only in the development of novel prevention and therapeutic strategies, but also help in better prediction of therapeutic responses in patients. This review summarizes some of the significant findings on the role of inflammation in breast cancer to gain collective molecular and mechanistic insights. We also discuss ongoing efforts and future outlook to exploit the existing knowledge for improved breast cancer management.
Collapse
Affiliation(s)
- Sachin Kumar Deshmukh
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA.,Department of Pathology, University of South Alabama, Mobile, AL, USA
| | - Sanjeev Kumar Srivastava
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA.,Department of Pathology, University of South Alabama, Mobile, AL, USA
| | - Teja Poosarla
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | - Donna Lynn Dyess
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | | | - Ajay Pratap Singh
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA.,Department of Pathology, University of South Alabama, Mobile, AL, USA.,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Seema Singh
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA.,Department of Pathology, University of South Alabama, Mobile, AL, USA.,Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
14510
|
Pardee TS, Luther S, Buyse M, Powell BL, Cortes J. Devimistat in combination with high dose cytarabine and mitoxantrone compared with high dose cytarabine and mitoxantrone in older patients with relapsed/refractory acute myeloid leukemia: ARMADA 2000 Phase III study. Future Oncol 2019; 15:3197-3208. [PMID: 31512500 DOI: 10.2217/fon-2019-0201] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Devimistat (CPI-613®) is an intravenously administered, novel lipoate analog that inhibits two key tricarboxcylic acid (TCA) cycle enzymes, pyruvate dehydrogenase (PDH) and α-ketoglutarate dehydrogenase complexes (KGDH). These complexes control TCA cycle entry of glucose and glutamine-derived carbons, respectively. Acute myeloid leukemia (AML) cells upregulate the TCA cycle in response to DNA damaging agents and treatment with devimistat increases sensitivity to them. A Phase I study of devimistat in combination with cytarabine and mitoxantrone produced a complete remission rate of 50% in patients with relapsed or refractory AML. In the combined Phase I/II experience, older patients with R/R AML treated with 2000 mg/m2 of devimistat had a 52% complete remission/complete remission with incomplete hematologic recovery rate and a median survival of 12.4 months. This report outlines the rationale and design of the ARMADA 2000 study, a Phase III clinical trial of devimistat in combination with high dose cytarabine and mitoxantrone compared with high dose cytarabine and mitoxantrone alone for older patients (≥60 years of age) with relapsed or refractory AML. Clinical trial registration: NCT#03504410.
Collapse
Affiliation(s)
- Timothy S Pardee
- Wake Forest Baptist Comprehensive Cancer Center, Department of Internal Medicine, Section on Hematology & Oncology, Winston-Salem, NC 27101, USA
- Rafael Pharmaceuticals, Cranbury, NJ 08512, USA
| | | | - Marc Buyse
- International Drug Development Institute, Louvain-La-Neuve, Belgium
| | - Bayard L Powell
- Wake Forest Baptist Comprehensive Cancer Center, Department of Internal Medicine, Section on Hematology & Oncology, Winston-Salem, NC 27101, USA
| | - Jorge Cortes
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
14511
|
Risk factors for lymph nodes involvement in obese women with endometrial carcinomas. Gynecol Oncol 2019; 155:27-33. [DOI: 10.1016/j.ygyno.2019.07.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/15/2019] [Accepted: 07/16/2019] [Indexed: 11/18/2022]
|
14512
|
Oh S, Ryu E. Does Holding Back Cancer-Related Concern Affect Couples' Marital Relationship and Quality of Life of Patients with Lung Cancer? An Actor–Partner Interdependence Mediation Modeling Approach. Asian Nurs Res (Korean Soc Nurs Sci) 2019; 13:277-285. [PMID: 31605768 DOI: 10.1016/j.anr.2019.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 11/15/2022] Open
Affiliation(s)
- Soonyoung Oh
- Department of Nursing, Kyungbok University, Pochun, Republic of Korea
| | - Eunjung Ryu
- Department of Nursing, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
14513
|
Liu Y, Ye X, Yu Y, Lu S. Prognostic significance of anaplastic lymphoma kinase rearrangement in patients with completely resected lung adenocarcinoma. J Thorac Dis 2019; 11:4258-4270. [PMID: 31737311 DOI: 10.21037/jtd.2019.09.65] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Reports of the prognostic significance of anaplastic lymphoma kinase (ALK) rearrangement in early stage lung adenocarcinoma have been contradictory. This study aimed to identify the associations of ALK rearrangement with clinicopathologic features and prognosis in patients with surgically resected stage I-IIIA lung adenocarcinoma. Methods Analysis of ALK status was performed by a fully-automated immunochemistry assay (with rabbit monoclonal Ventana D5F3 antibody) in tissue sections of 2,103 patients with surgically-resected stage I-IIIA lung adenocarcinoma. ALK positive patients were matched with negative patients in a 1:1 ratio using propensity score matching (PSM). Clinical outcomes were assessed by disease-free survival (DFS) and overall survival (OS) after surgery. Initial recurrence pattern was also investigated according to ALK status. Results Among 2,103 stage I-IIIA lung adenocarcinoma cases, 81 (3.9%) were ALK positive. ALK positivity was significantly associated with younger age (P<0.001), solid predominant adenocarcinoma (P<0.001), variants of invasive adenocarcinoma (P<0.001), higher frequency of pleura invasion (P=0.040), smaller tumor size (P=0.014), mediastinal lymph node involvement (N2; P<0.001) and later pathologic stage (IIIA; P=0.001). In the match cohort, ALK positivity was not associated with DFS [hazard ratio (HR), 0.58; 95% confidence interval (CI): 0.33-1.03, P=0.063] or OS (HR, 0.61; 95% CI: 0.22-1.67, P=0.334). Lymph node involvement (HR: 5.36, 95% CI, 3.01-9.65, P<0.001) and solid predominant adenocarcinoma subtype (HR, 2.02; 95% CI: 1.07-3.79; P=0.029) were the independent prognostic factors of inferior DFS, and lymph node involvement was the independent prognostic factors of worse OS (HR, 6.61; 95% CI: 2.43-17.94; P<0.001). ALK positive patients had a higher risk of developing tumor recurrence in liver (P=0.043). Conclusions ALK rearrangement was not an independent prognostic factor in stage I-IIIA lung adenocarcinoma patients but leaded to a higher risk of developing recurrence in liver.
Collapse
Affiliation(s)
- Yinglei Liu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xiangyun Ye
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yongfeng Yu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
14514
|
Bourbonné V, Pradier O, Schick U, Servagi-Vernat S. Cancer of the oesophagus and lymph nodes management in the neoadjuvant or definitive radiochemotherapy setting. Cancer Radiother 2019; 23:682-687. [DOI: 10.1016/j.canrad.2019.07.130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/03/2019] [Accepted: 07/04/2019] [Indexed: 12/12/2022]
|
14515
|
Chen D, Wang CY. Targeting cancer stem cells in squamous cell carcinoma. PRECISION CLINICAL MEDICINE 2019; 2:152-165. [PMID: 31598386 PMCID: PMC6770277 DOI: 10.1093/pcmedi/pbz016] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/14/2019] [Accepted: 08/14/2019] [Indexed: 12/24/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a highly aggressive tumor and the sixth
most common cancer worldwide. Current treatment strategies for HNSCC are surgery,
radiotherapy, chemotherapy, immunotherapy or combinatorial therapies. However, the overall
5-year survival rate of HNSCC patients remains at about 50%. Cancer stem cells (CSCs), a
small population among tumor cells, are able to self-renew and differentiate into
different tumor cell types in a hierarchical manner, similar to normal tissue. In HNSCC,
CSCs are proposed to be responsible for tumor initiation, progression, metastasis, drug
resistance, and recurrence. In this review, we discuss the molecular and cellular
characteristics of CSCs in HNSCC. We summarize current approaches used in the literature
for identification of HNSCC CSCs, and mechanisms required for CSC regulation. We also
highlight the role of CSCs in treatment failure and therapeutic targeting options for
eliminating CSCs in HNSCC.
Collapse
Affiliation(s)
- Demeng Chen
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, UCLA, Los Angeles, CA 90095, USA
| | - Cun-Yu Wang
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, UCLA, Los Angeles, CA 90095, USA.,Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, UCLA, Los Angeles, CA 90095, USA.,Jonsson Comprehensive Cancer Center and Broad Stem Cell Research Center, UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
14516
|
Daneshmand S. Advanced Testis Cancer. Eur Urol Focus 2019; 5:710-712. [PMID: 31563547 DOI: 10.1016/j.euf.2019.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 09/17/2019] [Indexed: 10/25/2022]
Abstract
Management of advanced testis cancer requires dedicated attention to the nuances of the disease to not only optimize cure but also ensure lifelong health. We are rapidly entering an era of precision medicine with novel biomarker discoveries that will undoubtedly change the treatment paradigm in both early- and late-stage disease.
Collapse
Affiliation(s)
- Siamak Daneshmand
- Institute of Urology, USC/Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
14517
|
Li M, Zou X, Xia T, Wang T, Liu P, Zhou X, Wang S, Zhu W. A five-miRNA panel in plasma was identified for breast cancer diagnosis. Cancer Med 2019; 8:7006-7017. [PMID: 31568692 PMCID: PMC6853814 DOI: 10.1002/cam4.2572] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 08/17/2019] [Accepted: 09/10/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BC) is one of the most common cancers in females. Since early detection can bring prognosis benefit, discovery of novel noninvasive biomarkers for BC diagnosis is in urgent need. In this four‐phase study, we profiled miRNA expression in plasma samples from a total of 257 BC patients and 257 normal controls (NCs). Exiqon miRNA qPCR panel was used to select candidate miRNAs in the screening phase which were further analyzed using qRT‐PCR in the following training, testing and external validation phases. Finally, we identified five plasma miRNAs (let‐7b‐5p, miR‐122‐5p, miR‐146b‐5p, miR‐210‐3p and miR‐215‐5p) whose expression levels were significantly different between BC patients and NCs. A 5‐miRNA panel in plasma with high sensitivity and specificity was then constructed to detect BC. The areas under the receiver‐operating characteristic curves (AUCs) of the panel were 0.683, 0.966, 0.978 for the training, testing and external validation sets, respectively. Expression of the identified miRNAs was further analyzed among 32 pairs of BC tissue and the adjacent normal tissue samples as well as plasma‐derived exosome samples from 32 BC patients vs 32 NCs. Let‐7b‐5p was contrarily down‐regulated in BC tissue. In exosomes samples, only miR‐122‐5p was significantly up‐regulated as in plasma for BC patients. In conclusion, we identified a 5‐miRNA plasma panel (let‐7b‐5p, miR‐122‐5p, miR‐146b‐5p, miR‐210‐3p and miR‐215‐5p) that could serve as a promising biomarker for BC detection.
Collapse
Affiliation(s)
- Minghui Li
- Department of Breast Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Xuan Zou
- Fudan University Shanghai Cancer Center, Shanghai, PR China
| | - Tiansong Xia
- Department of Breast Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Tongshan Wang
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Ping Liu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Xin Zhou
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Shui Wang
- Department of Breast Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Wei Zhu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China.,Department of Oncology and Radiotherapy, Nanjing Pukou Central Hospital, Nanjing, PR China
| |
Collapse
|
14518
|
Yan W, Hu H, Tang B. Advances Of Chimeric Antigen Receptor T Cell Therapy In Ovarian Cancer. Onco Targets Ther 2019; 12:8015-8022. [PMID: 31686857 PMCID: PMC6777428 DOI: 10.2147/ott.s203550] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 09/10/2019] [Indexed: 12/22/2022] Open
Abstract
Ovarian cancer, as a common gynecological tumor, is currently recognized as the most lethal gynecological malignancy. In addition to conventional treatment methods such as surgery, radiotherapy and chemotherapy, adoptive immunotherapy represented by modified immune cells also shows good curative effects and is becoming an important method in the treatment of ovarian cancer. Studies have shown that most cancer cells can avoid the recognition of the immune system, thus limiting the anticancer effect of immunotherapy. Chimeric antigen receptor T (CAR-T) cell technology has emerged and has good targeting, killing, proliferation and persistence. A large number of clinical trials also have shown that this technology has achieved great success in improving the quality of life and prolonging the survival time of patients with malignant hematological tumors. CAR-T cell technology has become a research hotspot for immunotherapy. This article mainly reviews various CAR-T cell treatments and their specific mechanisms in the field of ovarian cancer treatment to provide new ideas for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Wenying Yan
- Department of Gynecology, Wangjiang Hospital, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Hongmei Hu
- Department of Gynecology, Sichuan Maternal and Child Health Hospital, Chengdu, Sichuan Province, People's Republic of China
| | - Biao Tang
- Department of Gynecology, Sichuan Maternal and Child Health Hospital, Chengdu, Sichuan Province, People's Republic of China
| |
Collapse
|
14519
|
Sumitomo R, Hirai T, Fujita M, Murakami H, Otake Y, Huang CL. M2 tumor-associated macrophages promote tumor progression in non-small-cell lung cancer. Exp Ther Med 2019; 18:4490-4498. [PMID: 31777551 DOI: 10.3892/etm.2019.8068] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/19/2019] [Indexed: 12/24/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are key components of the tumor microenvironment that can be polarized into different phenotypes, including tumor-inhibiting M1 macrophages and tumor-promoting M2 macrophages. To elucidate the biological and clinical significance of M2 TAMs in non-small-cell lung cancer (NSCLC), a comprehensive clinical assessment of the tissue distribution of M2 TAMs was performed. The tissue distribution of M2 TAMs was retrospectively analyzed using CD163 immunohistochemistry in 160 consecutive patients who underwent NSCLC resection. Tumor proliferation was evaluated via the Ki-67 proliferation index. The results revealed that the stromal density of M2 TAMs was significantly associated with the C-reactive protein (CRP) level (P=0.0250), the Ki-67 proliferation index (P=0.0090) and invasive size (P=0.0285). Furthermore, the stromal M2 TAM density was significantly associated with tumor differentiation (P=0.0018), lymph node metastasis (P=0.0347) and pathological stage (P=0.0412). The alveolar M2 TAM density was also significantly associated with the CRP level (P=0.0309), invasive size (P<0.0001), tumor differentiation (P=0.0192), tumor status (P=0.0108) and pathological stage (P=0.0110). By contrast, no association was observed between islet M2 TAM density and the aforementioned biological and clinical factors. In regards to prognosis, disease-free survival rate was significantly lower in patients with stromal M2 TAM-high tumors (P=0.0270) and in those with alveolar M2 TAM-high tumors (P=0.0283). Furthermore, the overall survival rate was also significantly lower in patients with stromal M2 TAM-high tumors (P=0.0162) and in those with alveolar M2 TAM-high tumors (P=0.0225). Therefore, during NSCLC progression, M2 TAMs may induce tumor cell aggressiveness and proliferation and increase metastatic potential, resulting in a poor prognosis in patients with NSCLC.
Collapse
Affiliation(s)
- Ryota Sumitomo
- Department of Thoracic Surgery, Tazuke Kofukai Medical Research Institute, Kitano Hospital, Osaka 530-8480, Japan
| | - Tatsuya Hirai
- Department of Thoracic Surgery, Tazuke Kofukai Medical Research Institute, Kitano Hospital, Osaka 530-8480, Japan
| | - Masaaki Fujita
- Department of Clinical Immunology and Rheumatology, Tazuke Kofukai Medical Research Institute, Kitano Hospital, Osaka 530-8480, Japan
| | - Hiroaki Murakami
- Department of Thoracic Surgery, Tazuke Kofukai Medical Research Institute, Kitano Hospital, Osaka 530-8480, Japan
| | - Yosuke Otake
- Department of Thoracic Surgery, Tazuke Kofukai Medical Research Institute, Kitano Hospital, Osaka 530-8480, Japan
| | - Cheng-Long Huang
- Department of Thoracic Surgery, Tazuke Kofukai Medical Research Institute, Kitano Hospital, Osaka 530-8480, Japan
| |
Collapse
|
14520
|
Pretalli JB, Frontczak Franck S, Pazart L, Roux C, Amiot C. Development of Ovarian Tissue Autograft to Restore Ovarian Function: Protocol for a French Multicenter Cohort Study. JMIR Res Protoc 2019; 8:e12944. [PMID: 31573931 PMCID: PMC6802486 DOI: 10.2196/12944] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 06/24/2019] [Accepted: 07/21/2019] [Indexed: 12/13/2022] Open
Abstract
Background Sterility is a major late effect of radiotherapy and chemotherapy treatments. Iatrogenic sterility is often permanent and greatly impacts long-term quality of life. Ovarian tissue cryopreservation (OTC) performed before gonadotoxic treatments with subsequent autograft is a method of fertility preservation available for girls and women. Its application in prepubertal girls is of particular value as it is the only possible approach in this patient group. In addition, it does not require a delay in cancer therapy and no ovarian stimulation is needed. Objective The primary aim of this protocol is to help increase the implementation of ovarian tissue autografting in France. Knowledge is still lacking regarding the efficacy of ovarian transplantation in restoring ovarian function and regarding the safety of this procedure, especially the risk of cancer cell reseeding in certain types of cancer. A secondary aim of this study is to generate data to improve our understanding of these two essential aspects. Methods The DATOR (Development of Ovarian Tissue Autograft in Order to Restore Ovarian Function) study is ongoing in 17 university hospitals. The DATOR protocol includes the autograft of ovarian cortex fragments. Candidates are identified from an observational prospective cohort (called the Prospective Cohort of Patients Candidates for Ovarian Tissue Autograft [PERIDATOR]) of patients who have undergone OTC. Enrollment in the study is initiated at the patient’s request and must be validated by the center’s multidisciplinary team and by the study steering committee. The DATOR study begins with a total medical checkup. Ovarian tissue qualification and residual disease detection, if required, are performed. Results The study is ongoing. Currently, 38 patients have provided informed consent and have been entered into the DATOR study. Graft has been performed for 34 of these patients. An interim analysis was conducted on the first 25 patients for whom the period of at least 1 year posttransplantation was achieved. Out of these 25 patients, 11 women succeeded in becoming pregnant (pregnancy rate=44% [11/25]; delivery rate=40% [10/25]). Among these, 6 women conceived twice, and 1 pregnancy led to a miscarriage. Conclusions Our preliminary analysis appears to be coherent with the accumulating body of evidence indicating the potential utility of ovarian tissue autograft for patients with premature ovarian failure. All these elements justify the pursuit of our study. Trial Registration ClinicalTrials.gov NCT02846064; https://clinicaltrials.gov/ct2/show/NCT02846064 International Registered Report Identifier (IRRID) DERR1-10.2196/12944
Collapse
Affiliation(s)
- Jean-Baptiste Pretalli
- INSERM CIC 1431, Centre d'Investigation Clinique, Centre Hospitalier Universitaire de Besançon, Besançon, France.,Department of Reproductive Medicine and Biology, Cryobiology, University Hospital of Besançon, Besançon, France
| | - Sophie Frontczak Franck
- Department of Reproductive Medicine and Biology, Cryobiology, University Hospital of Besançon, Besançon, France
| | - Lionel Pazart
- INSERM CIC 1431, Centre d'Investigation Clinique, Centre Hospitalier Universitaire de Besançon, Besançon, France.,EA481 - Integrative and Clinical Neuroscience Laboratory, University Bourgogne Franche-Comté, Besançon, France
| | - Christophe Roux
- INSERM CIC 1431, Centre d'Investigation Clinique, Centre Hospitalier Universitaire de Besançon, Besançon, France.,Department of Reproductive Medicine and Biology, Cryobiology, University Hospital of Besançon, Besançon, France.,INSERM, Établissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, University Bourgogne Franche-Comté, Besançon, France
| | - Clotilde Amiot
- INSERM CIC 1431, Centre d'Investigation Clinique, Centre Hospitalier Universitaire de Besançon, Besançon, France.,Department of Reproductive Medicine and Biology, Cryobiology, University Hospital of Besançon, Besançon, France.,INSERM, Établissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, University Bourgogne Franche-Comté, Besançon, France
| | | |
Collapse
|
14521
|
Xie W, Liu J, Huang X, Wu G, Jeen F, Chen S, Zhang C, Yang W, Li C, Li Z, Ge L, Tang W. A nomogram to predict vascular invasion before resection of colorectal cancer. Oncol Lett 2019; 18:5785-5792. [PMID: 31788051 PMCID: PMC6865036 DOI: 10.3892/ol.2019.10937] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 07/26/2019] [Indexed: 02/06/2023] Open
Abstract
Vascular invasion (VI) is an important feature for systemic recurrence and an indicator for the application of adjuvant therapy in colorectal cancer (CRC). Preoperative knowledge of VI is important in determining whether adjuvant therapy is necessary, as well as the adequacy of surgical resection. In the present study, a predictive nomogram for VI in patients with CRC was constructed. The prediction model consisted of 664 eligible patients with CRC, who were divided into a training set (n=468) and a validation set (n=196). Data were collected between August 2013 and April 2018. The feature selection model was established using the least absolute shrinkage and selection operator regression model. Multivariable logistic regression analysis was used to construct the predictive nomogram. The performance of the nomogram was evaluated by calibration, discrimination and clinical usefulness. Differentiation, computed tomography (CT)-based on N stage (CT N stage), hemameba and tumor distance from the anus (cm) were integrated into the nomogram. The nomogram exhibited good discrimination, with an area under the curve (AUC) of 0.731 and good calibration. Application of the nomogram in the validation cohort showed acceptable discrimination, with an AUC of 0.710 and good calibration. Decision curve analysis revealed that the nomogram was clinically useful. These findings suggests, to the best of our knowledge, that this may be the first nomogram for individual preoperative prediction of VI in patients with CRC, which may promote preoperative optimization strategies for this selected group of patients.
Collapse
Affiliation(s)
- Weishun Xie
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China.,Guangxi Clinical Research Center for Colorectal Cancer, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jungang Liu
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China.,Guangxi Clinical Research Center for Colorectal Cancer, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaoliang Huang
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China.,Guangxi Clinical Research Center for Colorectal Cancer, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Guo Wu
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China.,Guangxi Clinical Research Center for Colorectal Cancer, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Franco Jeen
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China.,Guangxi Clinical Research Center for Colorectal Cancer, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Shaomei Chen
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China.,Guangxi Clinical Research Center for Colorectal Cancer, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chuqiao Zhang
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China.,Guangxi Clinical Research Center for Colorectal Cancer, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Wenkang Yang
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China.,Guangxi Clinical Research Center for Colorectal Cancer, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chan Li
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zhengtian Li
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China.,Guangxi Clinical Research Center for Colorectal Cancer, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Lianying Ge
- Guangxi Clinical Research Center for Colorectal Cancer, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China.,Department of Gynecologic Oncology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Weizhong Tang
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China.,Guangxi Clinical Research Center for Colorectal Cancer, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
14522
|
Shi Y, Li Y, Yan C, Su H, Ying K. Identification of key genes and evaluation of clinical outcomes in lung squamous cell carcinoma using integrated bioinformatics analysis. Oncol Lett 2019; 18:5859-5870. [PMID: 31788059 PMCID: PMC6865087 DOI: 10.3892/ol.2019.10933] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 09/02/2019] [Indexed: 12/26/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide. Despite progress in the treatment of non-small-cell lung cancer, there are limited treatment options for lung squamous cell carcinoma (LUSC), compared with lung adenocarcinoma. The present study investigated the disease mechanism of LUSC in order to identify key candidate genes for diagnosis and therapy. A total of three gene expression profiles (GSE19188, GSE21933 and GSE74706) were analyzed using GEO2R to identify common differentially expressed genes (DEGs). The DEGs were then investigated using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis. A protein-protein interaction (PPI) network was constructed via the Search Tool for the Retrieval of Interacting Genes/Proteins, and visualized using Cytoscape software. The expression levels of the hub genes identified using CytoHubba were validated using the University of California, Santa Cruz (UCSC) database and the Human Protein Atlas. A Kaplan-Meier curve and Gene Expression Profiling Interactive Analysis were then employed to evaluate the associated prognosis and clinical pathological stage of the hub genes. Furthermore, non-coding RNA regulatory networks were constructed using the Gene-Cloud Biotechnology information website. A total of 359 common DEGs (155 upregulated and 204 downregulated) were identified, which were predominantly enriched in 'mitotic nuclear division', 'cell division', 'cell cycle' and 'p53 signaling pathway'. The PPI network consisted of 257 nodes and 2,772 edges, and the most significant module consisted of 66 upregulated genes. A total of 19 hub genes exhibited elevated RNA levels, and 10 hub genes had elevated protein levels compared with normal lung tissues. The upregulation of five hub genes (CCNB1, CEP55, FOXM1, MKI67 and TYMS; defined in Table I) were significantly associated with poor overall survival and unfavorable clinical pathological stages. Various ncRNAs, such as C1orf220, LINC01561 and MGC39584, may also play important roles in hub-gene regulation. In conclusion, the present study provides further understanding of the pathogenesis of LUSC, and reveals CCNB1, CEP55, FOXM1, MKI67 and TYMS as potential biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Yangfeng Shi
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Yeping Li
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Chao Yan
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Hua Su
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Kejing Ying
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| |
Collapse
|
14523
|
Yan L, Raj P, Yao W, Ying H. Glucose Metabolism in Pancreatic Cancer. Cancers (Basel) 2019; 11:cancers11101460. [PMID: 31569510 PMCID: PMC6826406 DOI: 10.3390/cancers11101460] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/25/2019] [Accepted: 09/25/2019] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal cancers, with a five-year survival rate of around 5% to 8%. To date, very few available drugs have been successfully used to treat PDAC due to the poor understanding of the tumor-specific features. One of the hallmarks of pancreatic cancer cells is the deregulated cellular energetics characterized by the “Warburg effect”. It has been known for decades that cancer cells have a dramatically increased glycolytic flux even in the presence of oxygen and normal mitochondrial function. Glycolytic flux is the central carbon metabolism process in all cells, which not only produces adenosine triphosphate (ATP) but also provides biomass for anabolic processes that support cell proliferation. Expression levels of glucose transporters and rate-limiting enzymes regulate the rate of glycolytic flux. Intermediates that branch out from glycolysis are responsible for redox homeostasis, glycosylation, and biosynthesis. Beyond enhanced glycolytic flux, pancreatic cancer cells activate nutrient salvage pathways, which includes autophagy and micropinocytosis, from which the generated sugars, amino acids, and fatty acids are used to buffer the stresses induced by nutrient deprivation. Further, PDAC is characterized by extensive metabolic crosstalk between tumor cells and cells in the tumor microenvironment (TME). In this review, we will give an overview on recent progresses made in understanding glucose metabolism-related deregulations in PDAC.
Collapse
Affiliation(s)
- Liang Yan
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Priyank Raj
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Wantong Yao
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Haoqiang Ying
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
14524
|
Abstract
Cancer remains one of the most difficult to manage healthcare problems. The last two decades have been considered the golden age of cancer research, with major breakthroughs being announced on a regular basis. However, the major problem regarding cancer treatment is the incapability to selectively target cancer cells, with certain populations of tumors still remaining alive after treatment. The main focus of researchers is to develop treatments that are both effective and selective in targeting malignant cells. In this regard, bioavailability can be increased by overcoming the biological barriers encountered in the active agent’s pathway, creating carrier vehicles that have the ability to target malignant cells and effectively release the active agent. Since its appearance, nanomedicine has provided many answers to these challenges, but still, some expectations were not satisfied. In this review, we focused on the most recent developments in targeted drug delivery. Furthermore, a summary of different types of nanoparticles used to deliver active therapeutic agents in oncology is presented, along with details on the nanodrugs that were clinically approved by the Food and Drug Administration (FDA), until April 2019.
Collapse
|
14525
|
Liu X, Zhang X, Bi J, Li Z, Zhang Z, Kong C. Caspase recruitment domain family member 10 regulates carbamoyl phosphate synthase 1 and promotes cancer growth in bladder cancer cells. J Cell Mol Med 2019; 23:8128-8138. [PMID: 31565867 PMCID: PMC6850932 DOI: 10.1111/jcmm.14683] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 08/21/2019] [Accepted: 09/01/2019] [Indexed: 12/22/2022] Open
Abstract
Bladder cancer, which can be divided into non‐muscle‐invasive and muscle‐invasive bladder cancer, is the most common urinary cancer in the United States. Caspase recruitment domain family member 10 (CARD10), also named CARD‐containing MAGUK protein 3 (CARMA3), is a member of the CARMA family and may activate the nuclear factor kappa B (NF‐κB) pathway. We utilized RNA sequencing and metabolic mass spectrometry to identify the molecular and metabolic feature of CARD10. The signalling pathway of CARD10 was verified by Western blotting analysis and functional assays. RNA sequencing and metabolic mass spectrometry of CARD10 knockdown identified the metabolic enzyme carbamoyl phosphate synthase 1 (CPS1) in the urea cycle as the downstream gene regulated by CARD10. We confirmed that CARD10 affected cell proliferation and nucleotide metabolism through regulating CPS1. We indicated that CARD10 promote bladder cancer growth via CPS1 and maybe a potential therapeutic target in bladder cancer.
Collapse
Affiliation(s)
- Xi Liu
- Department of Urology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Xiaotong Zhang
- Department of Urology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Jianbin Bi
- Department of Urology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Zhenhua Li
- Department of Urology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Zhe Zhang
- Department of Urology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Chuize Kong
- Department of Urology, The First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
14526
|
Relevance of Neurotrophin Receptors CD271 and TrkC for Prognosis, Migration, and Proliferation in Head and Neck Squamous Cell Carcinoma. Cells 2019; 8:cells8101167. [PMID: 31569361 PMCID: PMC6830344 DOI: 10.3390/cells8101167] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/16/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide and often has a poor prognosis. The present study investigated the role of the low affinity nerve growth factor receptor CD271 as a putative therapy target in HNSCC. Neurotrophins that bind to CD271 also have a high affinity for the tropomyosin receptor kinase family (Trk), consisting of TrkA, TrkB, and TrkC, which must also be considered in addition to CD271. A retrospective study and functional in vitro cell line tests (migration assay and cell sorting) were conducted in order to evaluate the relevance of CD271 expression alone and with regard to Trk expression. CD271 and Trks were heterogeneously expressed in human HNSCC. The vast majority of tumors exhibited CD271 and TrkA, whereas only half of the tumors expressed TrkB and TrkC. High expression of CD271-positive cells predicted a bad clinical outcome of patients with HNSCC and was associated with distant metastases. However, the human carcinomas that also expressed TrkC had a reduced correlation with distant metastases and better survival rates. In vitro, CD271 expression marked a subpopulation with higher proliferation rates, but proliferation was lower in tumor cells that co-expressed CD271 and TrkC. The CD271 inhibitor LM11A 31 suppressed cell motility in vitro. However, neither TrkA nor TrkB expression were linked to prognosis or cell proliferation. We conclude that CD271 is a promising candidate that provides prognostic information for HNSCC and could be a putative target for HNSCC treatment.
Collapse
|
14527
|
Toriihara A, Nobashi T, Baratto L, Duan H, Moradi F, Park S, Hatami N, Aparici CM, Davidzon G, Iagaru A. Comparison of 3 Interpretation Criteria for 68Ga-PSMA11 PET Based on Inter- and Intrareader Agreement. J Nucl Med 2019; 61:533-539. [PMID: 31562226 DOI: 10.2967/jnumed.119.232504] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/27/2019] [Indexed: 11/16/2022] Open
Abstract
PET using radiolabeled prostate-specific membrane antigen (PSMA) is now being more widely adopted as a valuable tool to evaluate patients with prostate cancer (PC). Recently, 3 different criteria for interpretation of PSMA PET were published: the European Association of Nuclear Medicine (EANM) criteria, the Prostate Cancer Molecular Imaging Standardized Evaluation criteria, and the PSMA Reporting and Data System. We compared these 3 criteria in terms of interreader, intrareader, and intercriteria agreement. Methods: Data from 104 patients prospectively enrolled in research protocols at our institution were retrospectively reviewed. The cohort consisted of 2 groups: 47 patients (mean age, 64.2 y old) who underwent Glu-NH-CO-NH-Lys-(Ahx)-[68Ga(HBED-CC)] (68Ga-PSMA11) PET/MRI for initial staging of biopsy-proven intermediate- or high-risk PC, and 57 patients (mean age, 70.5 y old) who underwent 68Ga-PSMA11 PET/CT because of biochemically recurrent PC. Three nuclear medicine physicians independently evaluated all 68Ga-PSMA11 PET/MRI and PET/CT studies according to the 3 interpretation criteria. Two of them reevaluated all studies 6 mo later in the same manner and masked to the initial reading. The Gwet agreement coefficient was calculated to evaluate interreader, intrareader, and intercriteria agreement based on the following sites: local lesion (primary tumor or prostate bed after radical prostatectomy), lymph node metastases, and other metastases. Results: In the PET/MRI group, interreader, intrareader, and intercriteria agreement ranged from substantial to almost perfect for any site according to all 3 criteria. In the PET/CT group, interreader agreement ranged from substantial to almost perfect except for judgment of distant metastases based on the PSMA Reporting and Data System (Gwet agreement coefficient, 0.57; moderate agreement), in which the most frequent cause of disagreement was lung nodules. Intrareader agreement ranged from substantial to almost perfect for any site according to all 3 criteria. Intercriteria agreement for each site was also substantial to almost perfect. Conclusion: Although the 3 published criteria have good interreader and intrareader reproducibility in evaluating 68Ga-PSMA11 PET, there are some factors causing interreader disagreement. Further work is needed to address this issue.
Collapse
Affiliation(s)
- Akira Toriihara
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Tomomi Nobashi
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Lucia Baratto
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Heying Duan
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Farshad Moradi
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Sonya Park
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Negin Hatami
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Carina Mari Aparici
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Guido Davidzon
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| |
Collapse
|
14528
|
Turanli B, Altay O, Borén J, Turkez H, Nielsen J, Uhlen M, Arga KY, Mardinoglu A. Systems biology based drug repositioning for development of cancer therapy. Semin Cancer Biol 2019; 68:47-58. [PMID: 31568815 DOI: 10.1016/j.semcancer.2019.09.020] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/20/2023]
Abstract
Drug repositioning is a powerful method that can assists the conventional drug discovery process by using existing drugs for treatment of a disease rather than its original indication. The first examples of repurposed drugs were discovered serendipitously, however data accumulated by high-throughput screenings and advancements in computational biology methods have paved the way for rational drug repositioning methods. As chemotherapeutic agents have notorious side effects that significantly reduce quality of life, drug repositioning promises repurposed noncancer drugs with little or tolerable adverse effects for cancer patients. Here, we review current drug-related data types and databases including some examples of web-based drug repositioning tools. Next, we describe systems biology approaches to be used in drug repositioning for effective cancer therapy. Finally, we highlight examples of mostly repurposed drugs for cancer treatment and provide an overview of future expectations in the field for development of effective treatment strategies.
Collapse
Affiliation(s)
- Beste Turanli
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden; Department of Bioengineering, Marmara University, Istanbul, Turkey; Department of Bioengineering, Istanbul Medeniyet University, Istanbul, Turkey
| | - Ozlem Altay
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital Gothenburg, Sweden
| | - Hasan Turkez
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum 25240, Turkey
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Mathias Uhlen
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden
| | | | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, United Kingdom.
| |
Collapse
|
14529
|
Mendoza LK, Ashford JM, Willard VW, Clark KN, Martin-Elbahesh K, Hardy KK, Merchant TE, Jeha S, Wang F, Zhang H, Conklin HM. Social Functioning of Childhood Cancer Survivors after Computerized Cognitive Training: A Randomized Controlled Trial. CHILDREN-BASEL 2019; 6:children6100105. [PMID: 31569616 PMCID: PMC6826733 DOI: 10.3390/children6100105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 11/16/2022]
Abstract
Childhood cancer survivors are at risk for cognitive and social deficits. Previous findings indicate computerized cognitive training can result in an improvement of cognitive skills. The current objective was to investigate whether these cognitive gains generalize to social functioning benefits. Sixty-eight survivors of childhood cancer were randomly assigned to a computerized cognitive intervention (mean age 12.21 ± 2.47 years, 4.97 ± 3.02 years off-treatment) or waitlist control group (mean age 11.82 ± 2.42 years, 5.04 ± 2.41 years off-treatment). Conners 3 Parent and Self-Report forms were completed pre-intervention, immediately post-intervention and six-months post-intervention. Piecewise linear mixed-effects models indicated no significant differences in Peer Relations between groups at baseline and no difference in change between groups from pre- to immediate post-intervention or post- to six-months post-intervention (ps > 0.40). Baseline Family Relations problems were significantly elevated in the control group relative to the intervention group (p < 0.01), with a significantly greater decline from pre- to immediate post-intervention (p < 0.05) and no difference in change between groups from post- to six-months post-intervention (p > 0.80). The study results suggest cognitive gains from computerized training do not generalize to social functioning. Training focused on skill-based social processing (e.g., affect recognition) may be more efficacious.
Collapse
Affiliation(s)
| | - Jason M Ashford
- St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | | | - Kellie N Clark
- St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | | | | | | | - Sima Jeha
- St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Fang Wang
- St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Hui Zhang
- St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | | |
Collapse
|
14530
|
Durm G, Hanna N. The shifting paradigm of biomarker-driven care in advanced non-small cell lung cancer (NSCLC). Transl Lung Cancer Res 2019; 8:539-542. [PMID: 31555527 DOI: 10.21037/tlcr.2019.04.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Greg Durm
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| | - Nasser Hanna
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| |
Collapse
|
14531
|
Ashrafizadeh M, Ahmadi Z, Kotla NG, Afshar EG, Samarghandian S, Mandegary A, Pardakhty A, Mohammadinejad R, Sethi G. Nanoparticles Targeting STATs in Cancer Therapy. Cells 2019; 8:E1158. [PMID: 31569687 PMCID: PMC6829305 DOI: 10.3390/cells8101158] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/20/2019] [Accepted: 09/25/2019] [Indexed: 12/12/2022] Open
Abstract
Over the past decades, an increase in the incidence rate of cancer has been witnessed. Although many efforts have been made to manage and treat this life threatening condition, it is still one of the leading causes of death worldwide. Therefore, scientists have attempted to target molecular signaling pathways involved in cancer initiation and metastasis. It has been shown that signal transducers and activator of transcription (STAT) contributes to the progression of cancer cells. This important signaling pathway is associated with a number of biological processes including cell cycle, differentiation, proliferation and apoptosis. It appears that dysregulation of the STAT signaling pathway promotes the migration, viability and malignancy of various tumor cells. Hence, there have been many attempts to target the STAT signaling pathway. However, it seems that currently applied therapeutics may not be able to effectively modulate the STAT signaling pathway and suffer from a variety of drawbacks such as low bioavailability and lack of specific tumor targeting. In the present review, we demonstrate how nanocarriers can be successfully applied for encapsulation of STAT modulators in cancer therapy.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran.
| | - Zahra Ahmadi
- Department of Basic Science, Shoushtar Branch, Islamic Azad University, Shoushtar 6451741117, Iran.
| | - Niranjan G Kotla
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Newcastle, Galway H91 W2TY, Ireland.
| | - Elham Ghasemipour Afshar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7619813159, Iran.
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur 9318614139, Iran.
| | - Ali Mandegary
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7619813159, Iran.
| | - Abbas Pardakhty
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7619813159, Iran.
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7616911319, Iran.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| |
Collapse
|
14532
|
Wang WJ, Li R, Guo CA, Li HT, Yu JP, Wang J, Xu ZP, Chen WK, Ren ZJ, Tao PX, Zhang YN, Wang C, Liu HB. Systematic assessment of complications after robotic-assisted total versus distal gastrectomy for advanced gastric cancer: A retrospective propensity score-matched study using Clavien-Dindo classification. Int J Surg 2019; 71:140-148. [PMID: 31568844 DOI: 10.1016/j.ijsu.2019.09.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 07/11/2019] [Accepted: 09/24/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Despite increasing evidence demonstrated robot-assisted distal gastrectomy (RADG) is safe and feasible for the treatment of advanced gastric cancer (AGC), robot-assisted total gastrectomy (RATG) remains a challenging procedure due to its technical difficulties and possible postoperative complications (POCs). This study aimed to systematically evaluate POCs following RATG. METHODS Between January 2017 and January 2019, 319 AGC patients with pathological stage T2-4aN0-3M0 who underwent RADG or RATG were enrolled. POCs were stratified using the Clavien-Dindo classification. One-to-one propensity score matching was performed to reduce confounding differences. RESULTS After matching, 266 patients met the criteria for further analysis. Ultimately, 64 patients (24.1%) who developed POCs had 126 clinical manifestation events. Overall the POCs rate was significantly greater after RATG in comparison with RADG (29.3% vs. 18.8%; P = 0.045), and more major POCs (Clavien-Dindo grade ≥ IIIa) were observed in the RATG group (14.3% vs. 5.3%; P = 0.013). The POCs were then classified into local and systemic POCs. The rates of local POCs (35.3% vs. 19.5%; P = 0.004) and systemic POCs (24.8% vs. 15.0%; P = 0.046) were significantly higher in the RATG group than the RADG group. Subgroup analysis showed that the anastomotic leakage rate was higher after RATG (5.3% vs. 0.8%; P = 0.031), whereas the remaining POCs were similar between the two groups. Patients with higher POCs significantly had longer postoperative length of stay (R = 0.895, P = 0.003). Multivariate analysis confirmed age, extent of resection, and TNM stage were risk factors for all POCs. CONCLUSIONS These findings demonstrated that RATG is technically feasible and safe for treatment of AGC with acceptable morbidity and mortality rates. The POCs rate of RATG was higher than RADG, especially for anastomotic leakage. More effective anastomotic techniques are needed in RATG to prevent leakage.
Collapse
Affiliation(s)
- Wen-Jie Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, PR China; Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, PR China; Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, Lanzhou, 730050, PR China
| | - Rui Li
- Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, PR China; Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, PR China
| | - Chang-An Guo
- Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, PR China; Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, Lanzhou, 730050, PR China; Department of Emergency, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, PR China
| | - Hong-Tao Li
- Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, 730050, Gansu, PR China
| | - Jian-Ping Yu
- Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, 730050, Gansu, PR China
| | - Jing Wang
- Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, 730050, Gansu, PR China; Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, 730000, Gansu, PR China
| | - Zi-Peng Xu
- Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, PR China; Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, 730050, Gansu, PR China
| | - Wei-Kai Chen
- Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, 730050, Gansu, PR China; Clinical Medical College, Gansu University of Traditional Chinese Medicine, Lanzhou, 730000, Gansu, PR China
| | - Zhi-Jian Ren
- Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, PR China; Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, PR China
| | - Peng-Xian Tao
- Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, PR China; Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, PR China
| | - Ya-Nan Zhang
- Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, PR China; Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, PR China
| | - Chen Wang
- Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, PR China; Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, 730030, Gansu, PR China.
| | - Hong-Bin Liu
- Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, PR China; Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, 730050, Gansu, PR China.
| |
Collapse
|
14533
|
Maino Vieytes CA, Mondul AM, Li Z, Zarins KR, Wolf GT, Rozek LS, Arthur AE. Dietary Fiber, Whole Grains, and Head and Neck Cancer Prognosis: Findings from a Prospective Cohort Study. Nutrients 2019; 11:nu11102304. [PMID: 31569808 PMCID: PMC6835374 DOI: 10.3390/nu11102304] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/17/2019] [Accepted: 09/25/2019] [Indexed: 02/06/2023] Open
Abstract
No studies, to date, have examined the relationship between dietary fiber and recurrence or survival after head and neck cancer diagnosis. The aim of this study was to determine whether pretreatment intake of dietary fiber or whole grains predicted recurrence and survival outcomes in newly diagnosed head and neck cancer (HNC) patients. This was a prospective cohort study of 463 participants baring a new head and neck cancer diagnosis who were recruited into the study prior to the initiation of any cancer therapy. Baseline (pre-treatment) dietary and clinical data were measured upon entry into the study cohort. Clinical outcomes were ascertained at annual medical reviews. Cox proportional hazard models were fit to examine the relationships between dietary fiber and whole grain intakes with recurrence and survival. There were 112 recurrence events, 121 deaths, and 77 cancer-related deaths during the study period. Pretreatment dietary fiber intake was inversely associated with risk of all-cause mortality (hazard ratio (HR): 0.37, 95% confidence interval (CI): 0.14–0.95, ptrend = 0.04). No statistically significant associations between whole grains and prognostic outcomes were found. We conclude that higher dietary fiber intake, prior to the initiation of treatment, may prolong survival time, in those with a new HNC diagnosis.
Collapse
Affiliation(s)
- Christian A Maino Vieytes
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Alison M Mondul
- Department of Epidemiology, University of Michigan, Ann Arbor, MI 48103, USA.
| | - Zonggui Li
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Katie R Zarins
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48103, USA.
| | - Gregory T Wolf
- Department of Otolaryngology, University of Michigan, Ann Arbor, MI 48103, USA.
| | - Laura S Rozek
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48103, USA.
- Department of Otolaryngology, University of Michigan, Ann Arbor, MI 48103, USA.
| | - Anna E Arthur
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
- Carle Cancer Center, Carle Foundation Hospital, Urbana, IL 61801, USA.
| |
Collapse
|
14534
|
Fan X, Zhao Y. miR-451a inhibits cancer growth, epithelial-mesenchymal transition and induces apoptosis in papillary thyroid cancer by targeting PSMB8. J Cell Mol Med 2019; 23:8067-8075. [PMID: 31559672 PMCID: PMC6850967 DOI: 10.1111/jcmm.14673] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/30/2019] [Accepted: 08/18/2019] [Indexed: 12/18/2022] Open
Abstract
Despite the increasing incidence of papillary thyroid cancer in the past decade, the molecular mechanism underlying its progression remains unknown. Several studies have reported down-regulation of miR-451a or circular miR-451a in papillary thyroid cancer cell lines or patients. However, the underlying molecular mechanism remains unknown. In this study, we found that overexpression of miR-451a could inhibit proliferation, epithelial-mesenchymal transition and induce apoptosis in papillary thyroid cancer cells. Proteasome subunit beta type-8 was predicted to be a direct target of miR-451a and was validated with a luciferase reporter assay. Further functional assays showed that miR-451a could inhibit thyroid cancer progression by targeting proteasome subunit beta type-8.
Collapse
Affiliation(s)
- Xinlong Fan
- Second Ward of Head & Neck Surgery, Liaoning Cancer Hospital & Institute (Cancer Hospital of China Medical University), Shenyang, China
| | - Yuejiao Zhao
- Second Ward of Head & Neck Surgery, Liaoning Cancer Hospital & Institute (Cancer Hospital of China Medical University), Shenyang, China
| |
Collapse
|
14535
|
Yang D, Ma Y, Zhao P, Ma J, He C. Systematic screening of protein-coding gene expression identified HMMR as a potential independent indicator of unfavorable survival in patients with papillary muscle-invasive bladder cancer. Biomed Pharmacother 2019; 120:109433. [PMID: 31568988 DOI: 10.1016/j.biopha.2019.109433] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/02/2019] [Accepted: 09/02/2019] [Indexed: 12/20/2022] Open
Abstract
Papillary and non-papillary are two histological patterns of bladder carcinogenesis and are considered as dual-track oncogenic pathways, which have different genetic alterations. The TCGA-bladder cancer (BLCA) database contains clinicopathological, genomic and survival data from over 400 muscle-invasive bladder cancer patients. In this study, using data from this database, we performed a systematic screening of gene expression to identify the protein-coding gene that might have prognostic value in papillary and non-papillary muscle-invasive bladder cancer (MIBC). The data of patients with primary MIBC in TCGA-BLCA was acquired from the UCSC Xena project (http://xena.ucsc.edu) for re-analysis. By setting |log2 fold change|≥2 and adjusted p value <0.01 as the screening criteria, we found 751 significantly dysregulated genes, including 183 overexpressed and 568 downregulated genes. HMMR was identified as a potential prognostic marker with unique expression. Multivariate analysis showed that its expression was an independent prognostic indicator of shorter progression-free survival (PFS) (HR: 1.400, 95%CI: 1.021-1.920, p = 0.037) in the papillary subtype. ENST00000393915.8 and ENST00000358715.3, two transcripts that contain all 18 exons and encode the full length of HMMR, were significantly upregulated in cancer tissues compared with normal bladder tissues. None of the 17 CpG sites in its DNA locus was relevant to HMMR expression. 26/403 (6.5%) MIBC cases had HMMR gene-level amplification, which was associated with upregulated HMMR expression compared with the copy-neutral and deletion groups. Gene set enrichment analysis (GSEA) in papillary MIBC found that the high HMMR expression group was associated with upregulated genes enriched in multiple gene sets with well-established role in BC development, including G2M checkpoint, E2 F Targets, Myc Targets V1, Myc Targets V2 and Glycolysis. Based on these findings, we infer that HMMR expression might be a specific prognostic marker in terms of PFS in papillary MIBC. DNA amplification might be an important mechanism of its elevation.
Collapse
Affiliation(s)
- Dong Yang
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Yan Ma
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Pengcheng Zhao
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Jing Ma
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Chaohong He
- Department of Urology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
14536
|
Carmichael H, Cowan M, McIntyre R, Velopulos C. Disparities in colorectal cancer mortality for rural populations in the United States: Does screening matter? Am J Surg 2019; 219:988-992. [PMID: 31604486 DOI: 10.1016/j.amjsurg.2019.09.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/08/2019] [Accepted: 09/24/2019] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Rural compared to urban populations in the US have increased colorectal cancer (CRC) incidence with known disparity in screening rates and mortality. We hypothesize that rural-urban disparities are different at a regional level. METHODS We assessed screening rates according to the 2016 Behavioral Risk Factor Surveillance System guidelines using state and city-level data for county level estimates and correlating with county CRC mortality data from the National Cancer Institute. We used multivariable modeling to examine associations between rurality, screening rates, and mortality. RESULTS Highest screening rate states had the smallest urban-rural disparities; lowest screening rate states had the largest disparities. Percent screened and urban-rural classification correlated significantly with mortality. Rural counties experienced ∼5 more deaths per 100,000 population even controlling for screening rates. CONCLUSIONS National urban-rural disparities in CRC screening mask greater state/regional disparities, not fully explaining the urban-rural mortality gap. Other factors (i.e. access to care, treatment differences) must be considered.
Collapse
Affiliation(s)
| | - Michelle Cowan
- University of Colorado, 12631 E 17th Avenue, C305, United States
| | - Robert McIntyre
- University of Colorado, 12631 E 17th Avenue, C305, United States
| | | |
Collapse
|
14537
|
Zhang T, Karsh LI, Nissenblatt MJ, Canfield SE. Androgen Receptor Splice Variant, AR-V7, as a Biomarker of Resistance to Androgen Axis-Targeted Therapies in Advanced Prostate Cancer. Clin Genitourin Cancer 2019; 18:1-10. [PMID: 31653572 DOI: 10.1016/j.clgc.2019.09.015] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 12/12/2022]
Abstract
Many therapeutic options are now available for men with metastatic castration-resistant prostate cancer (mCRPC), including next-generation androgen receptor axis-targeted therapies (AATTs), immunotherapy, chemotherapy, and radioisotope therapies. No clear consensus has been reached for the optimal sequencing of treatments for patients with mCRPC, and few well-validated molecular markers exist to guide the treatment decisions for individual patients. The androgen receptor splice variant 7 (AR-V7), a splice variant of the androgen receptor mRNA resulting in the truncation of the ligand-binding domain, has emerged as a biomarker for resistance to AATT. AR-V7 expression in circulating tumor cells has been associated with poor outcomes in patients treated with second- and third-line AATTs. Clinically validated assays are now commercially available for the AR-V7 biomarker. In the present review of the current literature, we have summarized the biology of resistance to AATT, with a focus on the AR-V7; and the clinical studies that have validated AR-V7 expression as a strong independent predictor of a lack of clinical benefit from AATTs. Existing evidence has indicated that patients with AR-V7-positive mCRPC will have better outcomes if treated with taxane chemotherapy regimens rather than additional AATTs.
Collapse
Affiliation(s)
- Tian Zhang
- Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University School of Medicine, Durham, NC.
| | | | - Michael J Nissenblatt
- Department of Medicine, Regional Cancer Care Associates and Robert Wood Johnson University Medical School, East Brunswick, NJ
| | - Steven E Canfield
- Department of Surgery, University of Texas McGovern Medical School, Houston, TX
| |
Collapse
|
14538
|
Affiliation(s)
- Daniel F Hayes
- From the University of Michigan Rogel Cancer Center, Ann Arbor
| |
Collapse
|
14539
|
Sims TT, Colbert LE, Zheng J, Delgado Medrano AY, Hoffman KL, Ramondetta L, Jazaeri A, Jhingran A, Schmeler KM, Daniel CR, Klopp A. Gut microbial diversity and genus-level differences identified in cervical cancer patients versus healthy controls. Gynecol Oncol 2019; 155:237-244. [PMID: 31500892 DOI: 10.1016/j.ygyno.2019.09.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/02/2019] [Accepted: 09/02/2019] [Indexed: 12/18/2022]
Abstract
OBJECTIVES The aim of this study was to characterize variation in the gut microbiome of women with locally advanced cervical cancer and compare it to healthy controls. METHODS We characterized the 16S rDNA fecal microbiome in 42 cervical cancer patients and 46 healthy female controls. Shannon diversity index (SDI) was used to evaluate alpha (within sample) diversity. Beta (between sample) diversity was examined using principle coordinate analysis (PCoA) of unweighted Unifrac distances. Relative abundance of microbial taxa was compared between samples using Linear Discriminant Analysis Effect Size (LEfSe). RESULTS Within cervical cancer patients, bacterial alpha diversity was positively correlated with age (p = 0.22) but exhibited an inverse relationship in control subjects (p < 0.01). Alpha diversity was significantly higher in cervical cancer patients as compared to controls (p < 0.05), though stratification by age suggested this relationship was restricted to older women (>50 years; p < 0.01). Beta diversity (unweighted Unifrac; p < 0.01) also significantly differed between cervical cancer patients and controls. Based on age- and race-adjusted LEfSe analysis, multiple taxa significantly differed between cervical cancer patients and controls. Prevotella, Porphyromonas, and Dialister were significantly enriched in cervical cancer patients, while Bacteroides, Alistipes and members of the Lachnospiracea family were significantly enriched in healthy subjects. CONCLUSION Our study suggests differences in gut microbiota diversity and composition between cervical cancer patients and controls. Associations within the gut microbiome by age may reflect etiologic/clinical differences. These findings provide rationale for further study of the gut microbiome in cervical cancer.
Collapse
Affiliation(s)
- Travis T Sims
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America.
| | - Lauren E Colbert
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Jiali Zheng
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Andrea Y Delgado Medrano
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Kristi L Hoffman
- Department of Molecular Virology and Microbiology, Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, United States of America
| | - Lois Ramondetta
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Amir Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Anuja Jhingran
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Kathleen M Schmeler
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Carrie R Daniel
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Ann Klopp
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| |
Collapse
|
14540
|
Shi X, Wang S, Jasbi P, Turner C, Hrovat J, Wei Y, Liu J, Gu H. Database-Assisted Globally Optimized Targeted Mass Spectrometry (dGOT-MS): Broad and Reliable Metabolomics Analysis with Enhanced Identification. Anal Chem 2019; 91:13737-13745. [PMID: 31556994 DOI: 10.1021/acs.analchem.9b03107] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Xiaojian Shi
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Scottsdale, Arizona 85259, United States
| | - Shuai Wang
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Scottsdale, Arizona 85259, United States
- Department of Vascular Surgery, The First Hospital of Jilin University, Jilin University, Changchun, Jilin Province 130021, People’s Republic of China
| | - Paniz Jasbi
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Scottsdale, Arizona 85259, United States
| | - Cassidy Turner
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Scottsdale, Arizona 85259, United States
| | - Jonathan Hrovat
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Scottsdale, Arizona 85259, United States
| | - Yiping Wei
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Scottsdale, Arizona 85259, United States
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province 330006, People’s Republic of China
| | - Jingping Liu
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Scottsdale, Arizona 85259, United States
- Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, People’s Republic of China
| | - Haiwei Gu
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Scottsdale, Arizona 85259, United States
| |
Collapse
|
14541
|
HDAC3 Activity is Essential for Human Leukemic Cell Growth and the Expression of β-catenin, MYC, and WT1. Cancers (Basel) 2019; 11:cancers11101436. [PMID: 31561534 PMCID: PMC6826998 DOI: 10.3390/cancers11101436] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 09/11/2019] [Accepted: 09/20/2019] [Indexed: 01/23/2023] Open
Abstract
Therapy of acute myeloid leukemia (AML) is unsatisfactory. Histone deacetylase inhibitors (HDACi) are active against leukemic cells in vitro and in vivo. Clinical data suggest further testing of such epigenetic drugs and to identify mechanisms and markers for their efficacy. Primary and permanent AML cells were screened for viability, replication stress/DNA damage, and regrowth capacities after single exposures to the clinically used pan-HDACi panobinostat (LBH589), the class I HDACi entinostat/romidepsin (MS-275/FK228), the HDAC3 inhibitor RGFP966, the HDAC6 inhibitor marbostat-100, the non-steroidal anti-inflammatory drug (NSAID) indomethacin, and the replication stress inducer hydroxyurea (HU). Immunoblotting was used to test if HDACi modulate the leukemia-associated transcription factors β-catenin, Wilms tumor (WT1), and myelocytomatosis oncogene (MYC). RNAi was used to delineate how these factors interact. We show that LBH589, MS-275, FK228, RGFP966, and HU induce apoptosis, replication stress/DNA damage, and apoptotic fragmentation of β-catenin. Indomethacin destabilizes β-catenin and potentiates anti-proliferative effects of HDACi. HDACi attenuate WT1 and MYC caspase-dependently and -independently. Genetic experiments reveal a cross-regulation between MYC and WT1 and a regulation of β-catenin by WT1. In conclusion, reduced levels of β-catenin, MYC, and WT1 are molecular markers for the efficacy of HDACi. HDAC3 inhibition induces apoptosis and disrupts tumor-associated protein expression.
Collapse
|
14542
|
PLCB4 upregulation is associated with unfavorable prognosis in pediatric acute myeloid leukemia. Oncol Lett 2019; 18:6057-6065. [PMID: 31788080 PMCID: PMC6865073 DOI: 10.3892/ol.2019.10921] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023] Open
Abstract
Phospholipase C (PLC) is a membrane-associated enzyme that regulates several cellular behaviors including cell motility, growth, transformation and differentiation. PLC is involved in cancer migration, invasion and drug resistance. However, the expression status and prognostic role of PLCB4 in acute myeloid leukemia (AML) remain unclear. In the present study, the complete clinical and mRNA expression data of 285 pediatric patients with de novo AML were obtained from the Therapeutically Available Research to Generate Effective Treatments database. The association between PLCB4 expression and clinical and molecular features was explored. The expression of PLCB4 was significantly higher in patients with AML who relapsed compared with those with long-term complete remission. Patients with PLCB4 upregulation had significantly lower overall survival (OS) and event free survival (EFS) rate compared with those with low PLCB4 expression. Multivariate Cox's regression analyses demonstrated that high PLCB4 expression was an independent risk factor of adverse OS (P<0.01; HR, 2.081) and EFS (P<0.01; HR, 2.130). Following stratification analysis according to transplant status in cases of first complete remission, the patients with high expression of PLCB4 had significantly lower OS and EFS rate in the chemotherapy group, but not the stem cell transplant group. Furthermore, PLCB4-associated genes were identified using Spearman's rank correlation analysis. KEGG pathway analysis revealed that PLCB4 and its associated genes were mainly involved in three potential pathways, including the Rap1 signaling pathway. Overall, the findings of the present study suggest that increased PLCB4 expression is associated with poor clinical outcome in pediatric patients with AML, and thus may represent a potential prognostic biomarker and therapeutic target for AML.
Collapse
|
14543
|
Yan L, Alba M, Tabassum N, Voelcker NH. Micro‐ and Nanosystems for Advanced Transdermal Delivery. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900141] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Li Yan
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Victoria 3052 Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing Clayton Victoria 3168 Australia
| | - Maria Alba
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Victoria 3052 Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing Clayton Victoria 3168 Australia
| | - Nazia Tabassum
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Victoria 3052 Australia
- The University of Central Punjab Johar Town Lahore 54000 Pakistan
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutical Sciences Monash University Parkville Victoria 3052 Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing Clayton Victoria 3168 Australia
- Melbourne Centre for Nanofabrication Victorian Node of the Australian National Fabrication Facility Clayton Victoria 3168 Australia
| |
Collapse
|
14544
|
Toyoshima Y, Kitamura H, Xiang H, Ohno Y, Homma S, Kawamura H, Takahashi N, Kamiyama T, Tanino M, Taketomi A. IL6 Modulates the Immune Status of the Tumor Microenvironment to Facilitate Metastatic Colonization of Colorectal Cancer Cells. Cancer Immunol Res 2019; 7:1944-1957. [PMID: 31554639 DOI: 10.1158/2326-6066.cir-18-0766] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 06/11/2019] [Accepted: 09/19/2019] [Indexed: 11/16/2022]
Abstract
It is unknown as to how liver metastases are correlated with host immune status in colorectal cancer. In this study, we found that IL6, a proinflammatory cytokine produced in tumor-bearing states, promoted the metastatic colonization of colon cancer cells in association with dysfunctional antitumor immunity. In IL6-deficient mice, metastatic colonization of CT26 cells in the liver was reduced, and the antitumor effector function of CD8+ T cells, as well as IL12 production by CD11c+ dendritic cells, were augmented in vivo IL6-deficient mice exhibited enhanced IFN-AR1-mediated type I interferon signaling, which upregulated PD-L1 and MHC class I expression on CT26 cells. In vivo injection of anti-PD-L1 effectively suppressed the metastatic colonization of CT26 cells in Il6 -/- but not in Il6 +/+ mice. Finally, we confirmed that colorectal cancer patients with low IL6 expression in their primary tumors showed prolonged disease-free survival. These findings suggest that IL6 may be a promising target for the treatment of metastasis in colorectal cancers by improving host immunity.
Collapse
Affiliation(s)
- Yujiro Toyoshima
- Division of Functional Immunology, Section of Disease Control, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hidemitsu Kitamura
- Division of Functional Immunology, Section of Disease Control, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.
| | - Huihui Xiang
- Division of Functional Immunology, Section of Disease Control, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.,Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yosuke Ohno
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shigenori Homma
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hideki Kawamura
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Norihiko Takahashi
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Toshiya Kamiyama
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Mishie Tanino
- Department of Surgical Pathology, Asahikawa Medical University, Asahikawa, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
14545
|
Zhang T, Bai R, Wang Q, Wang K, Li X, Liu K, Ryu J, Wang T, Chang X, Ma W, Bode AM, Xia Q, Song Y, Dong Z. Fluvastatin Inhibits HMG-CoA Reductase and Prevents Non-Small Cell Lung Carcinogenesis. Cancer Prev Res (Phila) 2019; 12:837-848. [PMID: 31554629 DOI: 10.1158/1940-6207.capr-19-0211] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/06/2019] [Accepted: 09/18/2019] [Indexed: 11/16/2022]
Abstract
Lung cancer is the leading cause of cancer-related death worldwide. However, promising agents for lung cancer prevention are still very limited. Identification of preventive targets and novel effective preventive agents is urgently needed for clinical applications. In this study, we found that fluvastatin targeted 3-Hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase (HMGCR), which a rate-limiting enzyme in the mevalonate pathway, and inhibited non-small cell lung cancer (NSCLC) tumorigenesis. Initially, we demonstrated that HMGCR is overexpressed in human lung adenocarcinoma tissues compared with normal tissues. Knockdown of HMGCR in NSCLC cells attenuated growth and induced apoptosis in vitro and in vivo Furthermore, we found that fluvastatin, an inhibitor of HMGCR, suppressed NSCLC cell growth and induced apoptosis. Intriguingly, fluvastastin functions by inhibiting the HMGCR-driven Braf/MEK/ERK1/2 and Akt signaling pathways. Notably, fluvastatin attenuated tumor growth in 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced lung tumorigenesis and in a patient-derived xenograft lung tumor model. Overall, our findings suggest that fluvastatin might be promising chemopreventive or potential therapeutic drug against NSCLC tumorigenesis, providing hope for rapid clinical translation.
Collapse
Affiliation(s)
- Tianshun Zhang
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Ruihua Bai
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qiushi Wang
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Keke Wang
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Xiang Li
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Kangdong Liu
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Joohyun Ryu
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Ting Wang
- The Hormel Institute, University of Minnesota, Austin, Minnesota.,China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| | - Xiaoyu Chang
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Weiya Ma
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Qingxin Xia
- Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yongping Song
- Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, Minnesota. .,China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, China
| |
Collapse
|
14546
|
Rawat M, Kadian K, Gupta Y, Kumar A, Chain PSG, Kovbasnjuk O, Kumar S, Parasher G. MicroRNA in Pancreatic Cancer: From Biology to Therapeutic Potential. Genes (Basel) 2019; 10:752. [PMID: 31557962 PMCID: PMC6827136 DOI: 10.3390/genes10100752] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 12/20/2022] Open
Abstract
Pancreatic cancer is one of the most aggressive malignancies, accounting for more than 45,750 deaths annually in the U.S. alone. The aggressive nature and late diagnosis of pancreatic cancer, coupled with the limitations of existing chemotherapy, present the pressing need for the development of novel therapeutic strategies. Recent reports have demonstrated a critical role of microRNAs (miRNAs) in the initiation, progression, and metastasis of cancer. Furthermore, aberrant expressions of miRNAs have often been associated with the cause and consequence of pancreatic cancer, emphasizing the possible use of miRNAs in the effective management of pancreatic cancer patients. In this review, we provide a brief overview of miRNA biogenesis and its role in fundamental cellular process and miRNA studies in pancreatic cancer patients and animal models. Subsequent sections narrate the role of miRNA in, (i) cell cycle and proliferation; (ii) apoptosis; (iii) invasions and metastasis; and (iv) various cellular signaling pathways. We also describe the role of miRNA's in pancreatic cancer; (i) diagnosis; (ii) prognosis and (iii) therapeutic intervention. Conclusion section describes the gist of review with future directions.
Collapse
Affiliation(s)
- Manmeet Rawat
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA.
| | - Kavita Kadian
- Department of Biotechnology, Kumaun University, Nainital, Uttarakhand 263001, India.
| | - Yash Gupta
- Department of Internal Medicine, Loyola University Medical Center, Chicago, IL 60153, USA.
| | - Anand Kumar
- Biosecurity and Public Health Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA.
| | - Patrick S G Chain
- Biosecurity and Public Health Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA.
| | - Olga Kovbasnjuk
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA.
| | - Suneel Kumar
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| | - Gulshan Parasher
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA.
| |
Collapse
|
14547
|
Yang H, Liang SQ, Schmid RA, Peng RW. New Horizons in KRAS-Mutant Lung Cancer: Dawn After Darkness. Front Oncol 2019; 9:953. [PMID: 31612108 PMCID: PMC6773824 DOI: 10.3389/fonc.2019.00953] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/09/2019] [Indexed: 12/12/2022] Open
Abstract
In non-small cell lung cancer (NSCLC), the most frequent oncogenic mutation in western countries is KRAS, for which, however, there remains no clinically approved targeted therapies. Recent progress on high biological heterogeneity including diverse KRAS point mutations, varying dependence on mutant KRAS, wide spectrum of other co-occurring genetic alterations, as well as distinct cellular status across the epithelial-to-mesenchymal transition (EMT), has not only deepened our understanding about the pathobiology of KRAS-mutant NSCLC but also brought about unprecedented new hopes for precision treatment of patients. In this review, we provide an update on the most recent advances in KRAS-mutant lung cancer, with a focus on mechanistic insights into tumor heterogeneity, the potential clinic implications and new therapies on horizons tailored for KRAS-mutant lung cancer.
Collapse
Affiliation(s)
- Haitang Yang
- Department of General Thoracic Surgery, Department of BioMedical Research, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Shun-Qing Liang
- Department of General Thoracic Surgery, Department of BioMedical Research, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- University of Massachusetts Medical School, Worcester, MA, United States
| | - Ralph A. Schmid
- Department of General Thoracic Surgery, Department of BioMedical Research, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ren-Wang Peng
- Department of General Thoracic Surgery, Department of BioMedical Research, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
14548
|
Tun AM, Thein KZ, Thein WL, Guevara E. Checkpoint inhibitors plus chemotherapy for first-line treatment of advanced non-small cell lung cancer: a systematic review and meta-analysis of randomized controlled trials. Future Sci OA 2019; 5:FSO421. [PMID: 31608159 PMCID: PMC6787520 DOI: 10.2144/fsoa-2019-0081] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background: We conducted a meta-analysis to evaluate the efficacy and safety of upfront add-on immunotherapy for advanced non-small cell lung cancers (NSCLC). Methods: We performed a literature search on first-line chemotherapy ± immunotherapy in NSCLC. We utilized Revman version 5.3 to calculate the estimated pooled hazard ratio for overall survival (OS) and progression-free survival (PFS) and pooled risk ratio for objective response rate (ORR), all-grade and high-grade adverse events with 95% CI. Results: We analyzed 4322 patients. The pooled hazard ratios for OS, PFS and ORR were 0.74 (95% CI: 0.62–0.88; p = 0.0007), 0.62 (95% CI: 0.57–0.68; p = 0.00001) and 1.51 (95% CI: 1.3–1.74; p = 0.00001), respectively. The pooled risk ratios for all-grade and high-grade adverse events were 1.01 (95% CI: 0.99–1.03; p = 0.27) and 1.17 (95% CI: 1.07–1.28; p = 0.0006), respectively. Conclusion: Add-on immunotherapy significantly improves PFS, OS and ORR for the first-line treatment of advanced NSCLC with a reasonable safety profile. Lung cancer is the most frequent cancer and is the leading cause of cancer mortality worldwide – more than half of the patients presented at late-stage disease, which is associated with limited survival. To treat cancers, we use immune checkpoint inhibitors that release the brakes on the immune system; thus, the immune cells can kill cancer cells better. Multiple clinical trials have tested the role of immune checkpoint inhibitors combined with chemotherapy for lung cancer treatment. Based on these clinical trials, we conducted a systematic review that showed improvement in outcomes with combined chemotherapy and immunotherapy with acceptable adverse events.
Collapse
Affiliation(s)
- Aung Myint Tun
- Department of Medicine, Division of Hematology & Oncology, The Brooklyn Hospital Center, Brooklyn, NY 11201, USA
| | - Kyaw Zin Thein
- Department of Hematology & Oncology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | | | - Elizabeth Guevara
- Department of Medicine, Division of Hematology & Oncology, The Brooklyn Hospital Center, Brooklyn, NY 11201, USA
| |
Collapse
|
14549
|
Zhao T, Qian K, Zhang Y. High Expression of FGF5 Is an Independent Prognostic Factor for Poor Overall Survival and Relapse-Free Survival in Lung Adenocarcinoma. J Comput Biol 2019; 27:948-957. [PMID: 31553229 DOI: 10.1089/cmb.2019.0241] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Lung cancer is not only a serious disease but also a public problem threatening human health all over the world. Nonsmall cell lung cancer-which accounts for the majority of lung cancer-is mainly composed of lung adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC). FGF5 is a gene located in q21.21. In the past years, research on FGF5 is mainly focused on hair length and hereditary spherocytosis. In our study, bioinformatics analysis of FGF5 was performed through multiple databases. Expression of FGF5 was compared between tumor and normal tissues, association between gene expression and clinical outcomes was investigated in LUAD and LUSC separately, and potential signaling pathways were predicted. FGF5 expression was upregulated in lung cancer tissues compared with normal tissues. What is more, the high FGF5 expression group had significantly lower proportions of lymph node negative (N0) patients (77/144, 53.5%, vs. 253/358, 70.7%, p = 0.000), and is associated with worse overall survival (OS) (p < 0.0001) and relapse-free survival (RFS) (p = 0.024) in LUAD patients, which could not be seen in LUSC. The following analysis confirmed that high FGF5 expression could be an independent prognostic factor for poor OS (HR: 0.431, 95% CI: 0.312-0.597, p = 0.001) and RFS (HR: 0.678, 95% CI: 0.471-0.977, p = 0.037) in LUAD, but not in LUSC. Coexpression genes related to FGF5 were explored and potential pathways were investigated for further research. FGF5 is a tumor-associated gene that upregulated in lung cancer tissues, and could be an independent prognostic factor that have potential value for further research.
Collapse
Affiliation(s)
- Teng Zhao
- Department of Thoracic Surgery, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Kun Qian
- Department of Thoracic Surgery, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Yi Zhang
- Department of Thoracic Surgery, Xuanwu Hospital Capital Medical University, Beijing, China
| |
Collapse
|
14550
|
Chen L, Qian K, Guo K, Zheng X, Sun W, Sun T, Wang Y, Li D, Wu Y, Ji Q, Wang Z. A Novel N Staging System for Predicting Survival in Patients with Medullary Thyroid Cancer. Ann Surg Oncol 2019; 26:4430-4438. [DOI: 10.1245/s10434-019-07871-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Indexed: 12/13/2022]
|