1
|
Underwood PW, Pawlik TM. Precision Medicine for Metastatic Colorectal Cancer: Where Do We Stand? Cancers (Basel) 2024; 16:3870. [PMID: 39594824 PMCID: PMC11593240 DOI: 10.3390/cancers16223870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/30/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Metastatic colorectal cancer is a leading cause of cancer-related death across the world. The treatment paradigm has shifted away from systemic chemotherapy alone to include targeted therapy and immunotherapy. The past two decades have been characterized by increased investigation into molecular profiling of colorectal cancer. These molecular profiles help physicians to better understand colorectal cancer biology among patients with metastatic disease. Additionally, improved data on genetic pathways allow for specific therapies to be targeted at the underlying molecular profile. Investigation of the EGFR, VEGF, HER2, and other pathways, as well as deficient mismatch repair, has led to the development of multiple targeted therapies that are now utilized in the National Comprehensive Cancer Network guidelines for colon and rectal cancer. While these new therapies have contributed to improved survival for metastatic colorectal cancer, long-term survival remains poor. Additional investigation to understand resistance to targeted therapy and development of new targeted therapy is necessary. New therapies are under development and are being tested in the preclinical and clinical settings. The aim of this review is to provide a comprehensive evaluation of molecular profiling, currently available therapies, and ongoing obstacles in the field of colorectal cancer.
Collapse
Affiliation(s)
| | - Timothy M. Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center, 395 W. 12th Ave., Suite 670, Columbus, OH 43210, USA;
| |
Collapse
|
2
|
Liu Y, Zhou Y, Chen P. Lung cancer organoids: models for preclinical research and precision medicine. Front Oncol 2023; 13:1293441. [PMID: 37941550 PMCID: PMC10628480 DOI: 10.3389/fonc.2023.1293441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 11/10/2023] Open
Abstract
Lung cancer is a malignancy with high incidence and mortality rates globally, and it has a 5-year survival rate of only 10%-20%. The significant heterogeneity in clinical presentation, histological features, multi-omics findings, and drug sensitivity among different lung cancer patients necessitate the development of personalized treatment strategies. The current precision medicine for lung cancer, primarily based on pathological and genomic multi-omics testing, fails to meet the needs of patients with clinically refractory lung cancer. Lung cancer organoids (LCOs) are derived from tumor cells within tumor tissues and are generated through three-dimensional tissue culture, enabling them to faithfully recapitulate in vivo tumor characteristics and heterogeneity. The establishment of a series of LCOs biobanks offers promising platforms for efficient screening and identification of novel targets for anti-tumor drug discovery. Moreover, LCOs provide supplementary decision-making factors to enhance the current precision medicine for lung cancer, thereby addressing the limitations associated with pathology-guided approaches in managing refractory lung cancer. This article presents a comprehensive review on the construction methods and potential applications of LCOs in both preclinical and clinical research. It highlights the significance of LCOs in biomarker exploration, drug resistance investigation, target identification, clinical precision drug screening, as well as microfluidic technology-based high-throughput drug screening strategies. Additionally, it discusses the current limitations and future prospects of this field.
Collapse
Affiliation(s)
- Yajing Liu
- School of Pharmacy, Qingdao University, Qingdao, China
- Research and Development Department, NanoPeptide (Qingdao) Biotechnology Ltd., Qingdao, China
| | - Yanbing Zhou
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Pu Chen
- Research and Development Department, NanoPeptide (Qingdao) Biotechnology Ltd., Qingdao, China
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
3
|
Tang YL, Li DD, Duan JY, Sheng LM, Wang X. Resistance to targeted therapy in metastatic colorectal cancer: Current status and new developments. World J Gastroenterol 2023; 29:926-948. [PMID: 36844139 PMCID: PMC9950860 DOI: 10.3748/wjg.v29.i6.926] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/24/2022] [Accepted: 01/31/2023] [Indexed: 02/10/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most lethal and common malignancies in the world. Chemotherapy has been the conventional treatment for metastatic CRC (mCRC) patients. However, the effects of chemotherapy have been unsatisfactory. With the advent of targeted therapy, the survival of patients with CRC have been prolonged. Over the past 20 years, targeted therapy for CRC has achieved substantial progress. However, targeted therapy has the same challenge of drug resistance as chemotherapy. Consequently, exploring the resistance mechanism and finding strategies to address the resistance to targeted therapy, along with searching for novel effective regimens, is a constant challenge in the mCRC treatment, and it is also a hot research topic. In this review, we focus on the current status on resistance to existing targeted therapies in mCRC and discuss future developments.
Collapse
Affiliation(s)
- Yuan-Ling Tang
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Dan-Dan Li
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Jia-Yu Duan
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Lei-Ming Sheng
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Xin Wang
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
- Department of Abdominal Cancer, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
4
|
Afroj T, Mitsuhashi A, Ogino H, Saijo A, Otsuka K, Yoneda H, Tobiume M, Nguyen NT, Goto H, Koyama K, Sugimoto M, Kondoh O, Nokihara H, Nishioka Y. Blockade of PD-1/PD-L1 Pathway Enhances the Antigen-Presenting Capacity of Fibrocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:1204-1214. [PMID: 33504617 PMCID: PMC7939041 DOI: 10.4049/jimmunol.2000909] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023]
Abstract
Fibrocytes, a distinct population of collagen-producing, monocyte-derived cells, are involved in wound healing as well as fibrotic diseases. Recently, fibrocytes have been revealed to play a role in the tumor microenvironment, particularly under antiangiogenic therapy. In addition, combination cancer immunotherapy with immune checkpoint inhibitor and antiangiogenic agents have been developed for various cancers in the clinical setting, although the immunological background is not clear. In the current study, we aimed to determine the function of fibrocytes in tumor immunity induced by immune checkpoint inhibitor therapy. Human and murine fibrocytes were generated from PBMCs and lungs, respectively. The expression of costimulatory and inhibitory molecules on fibrocytes was examined by flow cytometry. The stimulation of CD8+ T cells by fibrocytes was examined in MLRs with a 3H-thymidine incorporation assay. Fibrocytes expressed CD80low and CD86high as a costimulatory molecule, and expressed PD-L1high, but not PD-L2, as a coinhibitory molecule. Without any stimulation, fibrocytes strongly enhanced the proliferation of CD8+ T cells in mice and humans. Treatment with anti-CD86 and -CD54 Abs inhibited the growth of CD8+ T cells induced by fibrocytes. Anti-PD-L1 Ab further enhanced the proliferation of CD8+ T cells, even in the OVA-specific MLR with OT-1Rag-/- mice. Importantly, fibrocytes derived from PBMCs of patients with lung adenocarcinoma or murine MC38 tumors augmented the proliferation of CD8+ T cells with PD-L1 blockade. These results suggest that fibrocytes infiltrating tumor sites may play a role in the antitumor immunity mediated by CD8+ T cells when the activity is further enhanced by PD-L1/PD-1 blockade.
Collapse
Affiliation(s)
- Tania Afroj
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Atsushi Mitsuhashi
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Hirokazu Ogino
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Atsuro Saijo
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Kenji Otsuka
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Hiroto Yoneda
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Makoto Tobiume
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Na Thi Nguyen
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Hisatsugu Goto
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Kazuya Koyama
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Masamichi Sugimoto
- Product Research Department, Kamakura Research Laboratories, Chugai Pharmaceutical, Co., Ltd., Kanagawa 247-8530, Japan
| | - Osamu Kondoh
- Product Research Department, Kamakura Research Laboratories, Chugai Pharmaceutical, Co., Ltd., Kanagawa 247-8530, Japan
| | - Hiroshi Nokihara
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| | - Yasuhiko Nishioka
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan; and
| |
Collapse
|
5
|
Xu R, Zhou X, Wang S, Trinkle C. Tumor organoid models in precision medicine and investigating cancer-stromal interactions. Pharmacol Ther 2020; 218:107668. [PMID: 32853629 DOI: 10.1016/j.pharmthera.2020.107668] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023]
Abstract
Tumor development and progression require chemical and mechanical cues derived from cellular and non-cellular components in the tumor microenvironment, including the extracellular matrix (ECM), cancer-associated fibroblasts (CAFs), endothelial cells, and immune cells. Therefore, it is crucial to develop tissue culture models that can mimic in vivo cancer cell-ECM and cancer-stromal cell interactions. Three-dimensional (3D) tumor culture models have been widely utilized to study cancer development and progression. A recent advance in 3D culture is the development of patient-derived tumor organoid (PDO) models from primary human cancer tissue. PDOs maintain the heterogeneity of the primary tumor, which makes them more relevant for identifying therapeutic targets and verifying drug response. Other significant advances include development of 3D co-culture assays to investigate cell-cell interactions and tissue/organ morphogenesis, and microfluidic technology that can be integrated into 3D culture to mimic vasculature and blood flow. These advances offer spatial and temporal insights into cancer cell-stromal interactions and represent novel techniques to study tumor progression and drug response. Here, we summarize the recent progress in 3D culture and tumor organoid models, and discuss future directions and the potential of utilizing these models to study cancer-stromal interactions and direct personalized treatment.
Collapse
Affiliation(s)
- Ren Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40508, USA.
| | - Xiaotao Zhou
- Markey Cancer Center, University of Kentucky, Lexington, KY 40508, USA
| | - Shike Wang
- Markey Cancer Center, University of Kentucky, Lexington, KY 40508, USA
| | - Christine Trinkle
- Department of Mechanical Engineering, University of Kentucky, Lexington, KY, 40506, USA
| |
Collapse
|
6
|
Ricci B, Tycksen E, Celik H, Belle JI, Fontana F, Civitelli R, Faccio R. Osterix-Cre marks distinct subsets of CD45- and CD45+ stromal populations in extra-skeletal tumors with pro-tumorigenic characteristics. eLife 2020; 9:e54659. [PMID: 32755539 PMCID: PMC7428306 DOI: 10.7554/elife.54659] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 07/27/2020] [Indexed: 01/08/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are a heterogeneous population of mesenchymal cells supporting tumor progression, whose origin remains to be fully elucidated. Osterix (Osx) is a marker of osteogenic differentiation, expressed in skeletal progenitor stem cells and bone-forming osteoblasts. We report Osx expression in CAFs and by using Osx-cre;TdTomato reporter mice we confirm the presence and pro-tumorigenic function of TdTOSX+ cells in extra-skeletal tumors. Surprisingly, only a minority of TdTOSX+ cells expresses fibroblast and osteogenic markers. The majority of TdTOSX+ cells express the hematopoietic marker CD45, have a genetic and phenotypic profile resembling that of tumor infiltrating myeloid and lymphoid populations, but with higher expression of lymphocytic immune suppressive genes. We find Osx transcript and Osx protein expression early during hematopoiesis, in subsets of hematopoietic stem cells and multipotent progenitor populations. Our results indicate that Osx marks distinct tumor promoting CD45- and CD45+ populations and challenge the dogma that Osx is expressed exclusively in cells of mesenchymal origin.
Collapse
Affiliation(s)
- Biancamaria Ricci
- Department of Orthopedics, Washington University School of MedicineSt. LouisUnited States
| | - Eric Tycksen
- Genome Technology Access Center, Department of Genetics, Washington University School of MedicineSt. LouisUnited States
| | - Hamza Celik
- Department of Medicine, Division of Oncology, Washington University School of MedicineSt. LouisUnited States
| | - Jad I Belle
- Department of Medicine, Division of Oncology, Washington University School of MedicineSt. LouisUnited States
| | - Francesca Fontana
- Department of Medicine, Division of Oncology, Washington University School of MedicineSt. LouisUnited States
| | - Roberto Civitelli
- Department of Medicine, Division of Bone and Mineral Diseases, Washington University School of MedicineSt. LouisUnited States
| | - Roberta Faccio
- Department of Orthopedics, Washington University School of MedicineSt. LouisUnited States
- Shriners Children HospitalSt. LouisUnited States
| |
Collapse
|
7
|
Abstract
Purpose This review highlights the roles of fibrocytes—their origin, markers, regulation and functions—including contributions to corneal wound healing and fibrosis. Methods Literature review. Results Peripheral blood fibroblast-like cells, called fibrocytes, are primarily generated as mature collagen-producing cells in the bone marrow. They are likely derived from the myeloid lineage, although the exact precursor remains unknown. Fibrocytes are identified by a combination of expressed markers, such as simultaneous expression of CD34 or CD45 or CD11b and collagen type I or collagen type III. Fibrocytes migrate into the wound from the blood where they participate in pathogen clearance, tissue regeneration, wound closure and angiogenesis. Transforming growth factor beta 1 (TGF-β1) and adiponectin induce expression of α-smooth muscle actin and extracellular matrix proteins through activation of Smad3 and adenosine monophosphate-activated protein kinase pathways, respectively. Fibrocytes are important contributors to the cornea wound healing response and there are several mechanisms through which fibrocytes contribute to fibrosis in the cornea and other organs, such as their differentiation into myofibroblasts, production of matrix metalloproteinase, secretion of tissue inhibitor of metalloproteinase, and release of TGF-β1. In some tissues, fibrocytes may also contribute to the basement membrane regeneration and to the resolution of fibrosis. Conclusions New methods that block fibrocyte generation, fibrocyte migration, and their differentiation into myofibroblasts, as well as their production of matrix metalloproteinases, tissue inhibitor of metalloproteinase, and TGF-β1, have therapeutic potential to reduce the accumulation of collagens, maintain tissue integrity and retard or prevent the development of fibrosis.
Collapse
|
8
|
Duan H, Li X, Chen Y, Wang Y, Li Z. LncRNA RHPN1-AS1 promoted cell proliferation, invasion and migration in cervical cancer via the modulation of miR-299-3p/FGF2 axis. Life Sci 2019; 239:116856. [PMID: 31525429 DOI: 10.1016/j.lfs.2019.116856] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 12/16/2022]
Abstract
AIMS This study aims to determine the biological function and underlying mechanisms of lncRNA RHPN1 antisense RNA1 (RHPN1-AS1) in cervical cancer cell proliferation, invasion and migration. MAIN METHODS Gene expression was analysed by quantitative real-time PCR; protein levels were determined by western blot assay; in vitro functional assays determined the cervical cancer cell progression; in vivo tumor growth of cervical cancer cell was determined in nude mice xenograft models. KEY FINDINGS The results showed that RHPN1-AS1 was up-regulated in cervical cancer tissues and cell lines. In vitro functional assays demonstrated that RHPN1-AS1 overexpression promoted SiHa cell proliferation, invasion and migration; while RHPN1-AS1 knockdown showed the opposite effects. In vivo study showed that RHPN1-AS1 knockdown suppressed tumor growth in the nude mice. Further investigation showed that miR-299-3p was targeted and inversely regulated by RHPN1-AS1. In addition, miR-299-3p targeted the 3' untranslated region of fibroblast growth factor 2 (FGF2) to suppress its expression. The rescue experiments showed that the enhanced effects of RHPN1-AS1 overexpression on cell proliferation, growth, invasion and migration in SiHa cells were significantly attenuated by miR-299-3p overexpression or FGF2 inhibition. On the other hand, knockdown of miR-299-3p and overexpression of FGF2 both significantly increased cell proliferation, growth, invasion and migration in SiHa cells transfected with RHPN1-AS1 siRNA. SIGNIFICANCE In conclusion, our results revealed that RHPN1-AS1 promoted cervical cancer progression via targeting miR-299-3p/FGF2 axis. Our data suggested that RHPN1-AS1/miR-299-3p/FGF2 axis may be a promising target for cervical cancer treatment.
Collapse
Affiliation(s)
- Haixia Duan
- The Assisted Reproduction Gynaecology, Northwest Women and Children's Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Xiaojuan Li
- The Assisted Reproduction Gynaecology, Northwest Women and Children's Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Youyi Chen
- Department of Reproductive Center, Xi'an No.4 Hospital, Xi'an, China.
| | - Yan Wang
- Department of Obstetrics and Gynaecology, Shaanxi Weinan Central Hospital, Weinan, China.
| | - Zhibin Li
- Deparment of Obstetrics, Northwest Women and Children's Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
9
|
CXCL9/CXCL10 angiostasis CXC-chemokines in parallel with the CXCL12 as an angiogenesis CXC-chemokine are variously expressed in pre-eclamptic-women and their neonates. Pregnancy Hypertens 2019; 17:36-42. [DOI: 10.1016/j.preghy.2019.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 04/14/2019] [Accepted: 05/02/2019] [Indexed: 12/31/2022]
|
10
|
Chandler C, Liu T, Buckanovich R, Coffman LG. The double edge sword of fibrosis in cancer. Transl Res 2019; 209:55-67. [PMID: 30871956 PMCID: PMC6545239 DOI: 10.1016/j.trsl.2019.02.006] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/12/2019] [Accepted: 02/15/2019] [Indexed: 02/07/2023]
Abstract
Cancer-associated fibrosis is a critical component of the tumor microenvironment (TME) which significantly impacts cancer behavior. However, there is significant controversy regarding fibrosis as a predominantly tumor promoting or tumor suppressing factor. Cells essential to the generation of tissue fibrosis such as fibroblasts and mesenchymal stem cells (MSCs) have dual phenotypes dependent upon their independence or association with cancer cells. Cancer-associated fibroblasts and cancer-associated MSCs have unique molecular profiles which facilitate cancer cell cross talk, influence extracellular matrix deposition, and direct the immune system to generate a protumorigenic environment. In contrast, normal tissue fibroblasts and MSCs are important in restraining cancer initiation, influencing epithelial cell differentiation, and limiting cancer cell invasion. We propose this apparent dichotomy of function is due to (1) cancer mediated stromal reprogramming; (2) tissue stromal source; (3) unique subtypes of fibrosis; and (4) the impact of fibrosis on other TME elements. First, as cancer progresses, tumor cells influence their surrounding stroma to move from a cancer restraining phenotype into a cancer supportive role. Second, cancer has specific organ tropism, thus stroma derived from preferred metastatic organs support growth while less preferred metastatic tissues do not. Third, there are subtypes of fibrosis which have unique function to support or inhibit cancer growth. Fourth, depleting fibrosis influences other TME components which drive the cancer response. Collectively, this review highlights the complexity of cancer-associated fibrosis and supports a dual function of fibrosis which evolves during the continuum of cancer growth.
Collapse
Affiliation(s)
- Chelsea Chandler
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Tianshi Liu
- Department of Internal Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ronald Buckanovich
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Pittsburgh, Pittsburgh, Pennsylvania; Division of Hematology Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lan G Coffman
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Pittsburgh, Pittsburgh, Pennsylvania; Division of Hematology Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|