1
|
Quintanilha JCF, Sibley AB, Liu Y, Niedzwiecki D, Halabi S, Rogers L, O'Neil B, Kindler H, Kelly W, Venook A, McLeod HL, Ratain MJ, Nixon AB, Innocenti F, Owzar K. Common variation in a long non-coding RNA gene modulates variation of circulating TGF-β2 levels in metastatic colorectal cancer patients (Alliance). BMC Genomics 2024; 25:473. [PMID: 38745123 PMCID: PMC11092225 DOI: 10.1186/s12864-024-10354-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/25/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Herein, we report results from a genome-wide study conducted to identify protein quantitative trait loci (pQTL) for circulating angiogenic and inflammatory protein markers in patients with metastatic colorectal cancer (mCRC). The study was conducted using genotype, protein marker, and baseline clinical and demographic data from CALGB/SWOG 80405 (Alliance), a randomized phase III study designed to assess outcomes of adding VEGF or EGFR inhibitors to systemic chemotherapy in mCRC patients. Germline DNA derived from blood was genotyped on whole-genome array platforms. The abundance of protein markers was quantified using a multiplex enzyme-linked immunosorbent assay from plasma derived from peripheral venous blood collected at baseline. A robust rank-based method was used to assess the statistical significance of each variant and protein pair against a strict genome-wide level. A given pQTL was tested for validation in two external datasets of prostate (CALGB 90401) and pancreatic cancer (CALGB 80303) patients. Bioinformatics analyses were conducted to further establish biological bases for these findings. RESULTS The final analysis was carried out based on data from 540,021 common typed genetic variants and 23 protein markers from 869 genetically estimated European patients with mCRC. Correcting for multiple testing, the analysis discovered a novel cis-pQTL in LINC02869, a long non-coding RNA gene, for circulating TGF-β2 levels (rs11118119; AAF = 0.11; P-value < 1.4e-14). This finding was validated in a cohort of 538 prostate cancer patients from CALGB 90401 (AAF = 0.10, P-value < 3.3e-25). The analysis also validated a cis-pQTL we had previously reported for VEGF-A in advanced pancreatic cancer, and additionally identified trans-pQTLs for VEGF-R3, and cis-pQTLs for CD73. CONCLUSIONS This study has provided evidence of a novel cis germline genetic variant that regulates circulating TGF-β2 levels in plasma of patients with advanced mCRC and prostate cancer. Moreover, the validation of previously identified pQTLs for VEGF-A, CD73, and VEGF-R3, potentiates the validity of these associations.
Collapse
Affiliation(s)
- Julia C F Quintanilha
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Yingmiao Liu
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Donna Niedzwiecki
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
- Alliance Statistics and Data Management Center, Duke University, Durham, NC, USA
| | - Susan Halabi
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
- Alliance Statistics and Data Management Center, Duke University, Durham, NC, USA
| | - Layne Rogers
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Bert O'Neil
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| | - Hedy Kindler
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - William Kelly
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Alan Venook
- Department of Medicine, University of California at San Francisco, San Francisco, CA, USA
| | - Howard L McLeod
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Utah Tech University, St George, UT, USA
| | - Mark J Ratain
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Andrew B Nixon
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Federico Innocenti
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kouros Owzar
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA.
- Alliance Statistics and Data Management Center, Duke University, Durham, NC, USA.
| |
Collapse
|
2
|
Nixon AB, Liu Y, Yang Q, Luo B, Starr MD, Brady JC, Kelly WK, Beltran H, Morris MJ, George DJ, Armstrong AJ, Halabi S. Prognostic and predictive analyses of circulating plasma biomarkers in men with metastatic castration resistant prostate cancer treated with docetaxel/prednisone with or without bevacizumab. Prostate Cancer Prostatic Dis 2024:10.1038/s41391-024-00794-3. [PMID: 38347114 DOI: 10.1038/s41391-024-00794-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/16/2024] [Indexed: 02/28/2024]
Abstract
BACKGROUND CALGB 90401 (Alliance) was a phase III trial of 1050 patients with metastatic castration-resistant prostate cancer (mCRPC) comparing docetaxel, prednisone, bevacizumab (DP+B) versus DP alone. While this trial did not show an improvement in overall survival (OS), there were improved intermediate outcomes suggesting that subsets of men may derive benefit from this combination. The purpose of this analysis was to identify prognostic and predictive biomarkers associated with OS and progression-free survival (PFS) benefit from DP+B. METHODS Baseline EDTA plasma samples from 650 consenting patients were analyzed for 24 biomarkers. The proportional hazards model was utilized to test for the prognostic and predictive importance of the biomarkers for OS. The statistically significant biomarkers of OS were further investigated for prognostic and predictive importance for other secondary outcomes. RESULTS 15 markers [ICAM-1, VEGF-R3, TIMP-1, TSP-2, Ang-2, Her-3, Osteopontin (OPN), PlGF, VCAM-1, HGF, VEGF, Chromogranin A, IL-6, VEGF-R1, BMP-9] were prognostic of OS, while 9 markers (ICAM-1, VEGF-R3, Her-3, TIMP-1, Ang-2, OPN, PlGF, HGF, and VEGF) were also prognostic of PFS. All markers were statistically significant in univariate analyses after adjustment for multiplicity (FDR < 0.1). In multivariable analyses of OS adjusting for risk score, seven markers had FDR < 0.1, including ICAM-1, VEGF-R3, TIMP-1, Ang-2, VEGF, TSP-2 and HGF. In unadjusted analysis, OPN was predictive of PFS improvement with DP+B, in both univariate and multivariable analysis. However, none of the biomarkers tested were predictive of clinical outcomes after adjusting for multiple comparisons. CONCLUSIONS Multiple biomarkers were identified in CALGB 90401 as prognostic of clinical outcomes but not predictive of OS. While OPN may have promise as a potential biomarker for anti-angiogenic therapies, further mechanistic and clinical studies are needed to determine the underlying biology and potential clinical application.
Collapse
Affiliation(s)
- Andrew B Nixon
- Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| | - Yingmiao Liu
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Qian Yang
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| | - Bin Luo
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| | - Mark D Starr
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - John C Brady
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Wm Kevin Kelly
- Division of Solid Tumor Oncology, Department of Medical Oncology and Urology, Thomas Jefferson University and Sidney Kimmel Cancer Center, Philadelphia, PA, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Michael J Morris
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel J George
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University Medical Center, Durham, NC, USA
| | - Andrew J Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University Medical Center, Durham, NC, USA
| | - Susan Halabi
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
3
|
Quintanilha JC, Sibley AB, Liu Y, Niedzwiecki D, Halabi S, Rogers L, O’Neil B, Kindler H, Kelly W, Venook A, McLeod HL, Ratain MJ, Nixon AB, Innocenti F, Owzar K. Common variation in a long non-coding RNA gene modulates variation of circulating TGF- β2 levels in metastatic colorectal cancer patients (Alliance). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.04.23298815. [PMID: 38106038 PMCID: PMC10723514 DOI: 10.1101/2023.12.04.23298815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Background Herein, we report results from a genome-wide study conducted to identify protein quantitative trait loci (pQTL) for circulating angiogenic and inflammatory protein markers in patients with metastatic colorectal cancer (mCRC).The study was conducted using genotype, protein marker, and baseline clinical and demographic data from CALGB/SWOG 80405 (Alliance), a randomized phase III study designed to assess outcomes of adding VEGF or EGFR inhibitors to systemic chemotherapy in mCRC patients. Germline DNA derived from blood was genotyped on whole-genome array platforms. The abundance of protein markers was quantified using a multiplex enzyme-linked immunosorbent assay from plasma derived from peripheral venous blood collected at baseline. A robust rank-based method was used to assess the statistical significance of each variant and protein pair against a strict genome-wide level. A given pQTL was tested for validation in two external datasets of prostate (CALGB 90401) and pancreatic cancer (CALGB 80303) patients. Bioinformatics analyses were conducted to further establish biological bases for these findings. Results The final analysis was carried out based on data from 540,021 common typed genetic variants and 23 protein markers from 869 genetically estimated European patients with mCRC. Correcting for multiple testing, the analysis discovered a novel cis-pQTL in LINC02869, a long non-coding RNA gene, for circulating TGF-β2 levels (rs11118119; AAF = 0.11; P-value < 1.4e-14). This finding was validated in a cohort of 538 prostate cancer patients from CALGB 90401 (AAF = 0.10, P-value < 3.3e-25). The analysis also validated a cis-pQTL we had previously reported for VEGF-A in advanced pancreatic cancer, and additionally identified trans-pQTLs for VEGF-R3, and cis-pQTLs for CD73. Conclusions This study has provided evidence of a novel cis germline genetic variant that regulates circulating TGF-β2 levels in plasma of patients with advanced mCRC and prostate cancer. Moreover, the validation of previously identified pQTLs for VEGF-A, CD73, and VEGF-R3, potentiates the validity of these associations.
Collapse
Affiliation(s)
- Julia C.F. Quintanilha
- UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alexander B. Sibley
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Yingmiao Liu
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Donna Niedzwiecki
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
- Alliance Statistics and Data Management Center, Duke University, Durham, NC, USA
| | - Susan Halabi
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
- Alliance Statistics and Data Management Center, Duke University, Durham, NC, USA
| | - Layne Rogers
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Bert O’Neil
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, USA
| | - Hedy Kindler
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - William Kelly
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Alan Venook
- Department of Medicine, University of California at San Francisco, San Francisco, California, USA
| | - Howard L. McLeod
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; and Utah Tech University, St George, UT, USA (current); and Intermountain Healthcare, St George, UT, USA (current)
| | - Mark J. Ratain
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Andrew B. Nixon
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Federico Innocenti
- Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kouros Owzar
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina, USA
- Alliance Statistics and Data Management Center, Duke University, Durham, NC, USA
| |
Collapse
|
4
|
Yuki S, Yamazaki K, Sunakawa Y, Taniguchi H, Bando H, Shiozawa M, Nishina T, Yasui H, Kagawa Y, Takahashi N, Denda T, Esaki T, Kawakami H, Satake H, Takashima A, Matsuhashi N, Kato T, Asano C, Abe Y, Nomura S, Yoshino T. Role of plasma angiogenesis factors in the efficacy of first-line chemotherapy combined with biologics in RAS wild-type metastatic colorectal cancer: Results from the GI-SCREEN CRC-Ukit study. Cancer Med 2023; 12:18702-18716. [PMID: 37641470 PMCID: PMC10557901 DOI: 10.1002/cam4.6486] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/08/2023] [Accepted: 08/19/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND Several biomarkers have been established for metastatic colorectal cancer (mCRC). We investigated whether plasma angiogenesis factors could predict the efficacy of biologics combined with chemotherapy in first-line (1L) treatment in patients with RAS wild-type mCRC and the dynamics of plasma angiogenesis factors at progression during 1L treatment. METHODS In this multicenter prospective observational study, serial plasma samples were prospectively collected at pretreatment and progression stages; 17 plasma angiogenesis factors were analyzed using the multiplex assay with Luminex® technology. Interactions between the pretreatment measurements and treatment groups on progression-free survival (PFS) and overall survival (OS) in patients with RAS wild-type were assessed using the propensity-score weighted Cox proportional hazards model. RESULTS From February 2018 to September 2020, 202 patients were enrolled in the 1L cohort; 133 patients had RAS wild-type (chemotherapy plus bevacizumab [BEV group, n = 33] and plus anti-epidermal growth factor receptor monoclonal antibodies [aEGFR group, n = 100]). A trend of strong interaction on PFS was observed for interleukin-8 (IL-8) (p = 0.0752) and soluble vascular cell adhesion molecule-1 (sVCAM-1) (p = 0.0156). Regarding OS, IL-8 (p = 0.0283), soluble vascular endothelial growth factor-receptor-1 (sVEGFR-1) (p = 0.0777) and sVCAM-1 (p = 0.0011) tended to differentiate the treatment effect. In 112 patients, plasma samples were evaluable for dynamic analysis (57 and 55 from the BEV and aEGFR groups, respectively). In the BEV group, six factors significantly increased during progression, whereas two decreased. In the aEGFR group, three factors significantly increased, and six decreased. CONCLUSION Pretreatment plasma IL-8 and sVCAM-1 levels could be predictive biomarkers to distinguish BEV and anti-EGFR mAbs when combined with chemotherapy in the 1L treatment of RAS wild-type mCRC. Several plasma angiogenesis factors showed significant change at progression in 1L chemotherapy plus biologics for RAS wild-type mCRC, which are potential biomarkers for selecting an optimal angiogenesis inhibitor in second-line treatment.
Collapse
Affiliation(s)
- Satoshi Yuki
- Department of Gastroenterology and HepatologyHokkaido University HospitalSapporoJapan
| | - Kentaro Yamazaki
- Division of Gastrointestinal OncologyShizuoka Cancer CenterNagaizumiJapan
| | - Yu Sunakawa
- Department of Clinical OncologySt. Marianna University School of MedicineKawasakiJapan
| | - Hiroya Taniguchi
- Department of Clinical OncologyAichi Cancer Center HospitalNagoyaJapan
| | - Hideaki Bando
- Department of Gastroenterology and Gastrointestinal OncologyNational Cancer Center Hospital EastKashiwaJapan
| | - Manabu Shiozawa
- Department of Gastrointestinal SurgeryKanagawa Cancer CenterYokohamaJapan
| | - Tomohiro Nishina
- Department of Gastrointestinal Medical OncologyNational Hospital Organization Shikoku Cancer CenterMatsuyamaJapan
| | - Hisateru Yasui
- Department of Medical OncologyKobe City Medical Center General HospitalKobeJapan
| | | | | | | | - Taito Esaki
- Department of Gastrointestinal and Medical OncologyNational Hospital Organization Kyushu Cancer CenterFukuokaJapan
| | - Hisato Kawakami
- Department of Medical OncologyKindai University Faculty of MedicineOsaka‐sayamaJapan
| | - Hironaga Satake
- Cancer Treatment CenterKansai Medical University HospitalHirakata
- Department of Medical OncologyKochi Medical SchoolNankokuJapan
| | - Atsuo Takashima
- Department of Gastrointestinal Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Nobuhisa Matsuhashi
- Department of Gastroenterological Surgery and Pediatric SurgeryGifu University HospitalGifuJapan
| | - Takeshi Kato
- Department of SurgeryNational Hospital Organization Osaka National HospitalOsakaJapan
| | - Chiharu Asano
- Institute of Health Science Innovation for Medical CareHokkaido University HospitalSapporoJapan
| | | | - Shogo Nomura
- Department of Biostatistics and Bioinformatics, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal OncologyNational Cancer Center Hospital EastKashiwaJapan
| |
Collapse
|
5
|
Harrington J, Nixon AB, Daubert MA, Yow E, Januzzi J, Fiuzat M, Whellan DJ, O'Connor CM, Ezekowitz J, Piña IL, Adams KF, Felker GM, Karra R. Circulating Angiokines Are Associated With Reverse Remodeling and Outcomes in Chronic Heart Failure. J Card Fail 2023; 29:896-906. [PMID: 36632934 PMCID: PMC10272021 DOI: 10.1016/j.cardfail.2022.12.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 01/09/2023]
Abstract
BACKGROUND We sought to determine whether circulating modifiers of endothelial function are associated with cardiac structure and clinical outcomes in patients with heart failure with reduced ejection fraction (HFrEF). METHODS We measured 25 proteins related to endothelial function in 99 patients from the GUIDE-IT study. Protein levels were evaluated for association with echocardiographic parameters and the incidence of all-cause death and hospitalization for heart failure (HHF). RESULTS Higher concentrations of angiopoietin 2 (ANGPT2), vascular endothelial growth factor receptor 1 (VEGFR1) and hepatocyte growth factor (HGF) were significantly associated with worse function and larger ventricular volumes. Over time, decreases in ANGPT2 and, to a lesser extent, VEGFR1 and HGF, were associated with improvements in cardiac size and function. Individuals with higher concentrations of ANGPT2, VEGFR1 or HGF had increased risks for a composite of death and HHF in the following year (HR 2.76 (95% CI 1.73-4.40) per 2-fold change in ANGPT2; HR 1.76 (95% CI 1.11-2.79) for VEGFR1; and HR 4.04 (95% CI 2.19-7.44) for HGF). CONCLUSIONS Proteins related to endothelial function associate with cardiac size, cardiac function and clinical outcomes in patients with HFrEF. These results support the concept that endothelial function may be an important contributor to the progression to and the recovery from HFrEF.
Collapse
Affiliation(s)
- Josephine Harrington
- Department of Medicine, Duke University Medical Center, Durham, NC; Duke Clinical Research Institute, Durham, NC
| | - Andrew B Nixon
- Department of Medicine, Duke University Medical Center, Durham, NC; Duke Cancer Institute, Duke University Medical Center, Durham, NC
| | - Melissa A Daubert
- Department of Medicine, Duke University Medical Center, Durham, NC; Duke Clinical Research Institute, Durham, NC
| | - Eric Yow
- Duke Clinical Research Institute, Durham, NC
| | - James Januzzi
- Massachusetts General Hospital; Harvard Medical School, Boston, MA; Baim Institute for Clinical Research, Boston, MA
| | - Mona Fiuzat
- Duke Clinical Research Institute, Durham, NC
| | - David J Whellan
- Department of Medicine, Thomas Jefferson University, Philadelphia, PA
| | | | - Justin Ezekowitz
- Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada
| | | | - Kirkwood F Adams
- Department of Medicine, University of North Carolina, Chapel Hill, NC
| | - G Michael Felker
- Department of Medicine, Duke University Medical Center, Durham, NC; Duke Clinical Research Institute, Durham, NC
| | - Ravi Karra
- Department of Medicine, Duke University Medical Center, Durham, NC; Department of Pathology, Duke University Medical Center, Durham, NC.
| |
Collapse
|
6
|
Long-term response of more than 9 years to regorafenib in a heavily pretreated patient with metastatic colorectal cancer. Anticancer Drugs 2023; 34:451-454. [PMID: 36730636 DOI: 10.1097/cad.0000000000001410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide, with approximately 1.9 million new diagnoses and 935 000 deaths annually. Overall, there is accumulating evidence that receiving all available treatments leads to a survival advantage and, although tailored treatments might be appropriate for selected patients, the one-size-fits-all approach is still widely used in chemo-refractory patients. Currently, different antiangiogenics and multitarget agents are indicated in treatment of metastatic CRC (mCRC) whereas the identification of useful predictive factors for the treatment response is lacking. Analysis of potential predictive biomarkers of efficacy of regorafenib is still ongoing but may prove to be difficult because of its nonspecific activity across a wide range of angiogenic, oncogenic, stromal, and intracellular signaling kinases. We present a case of a 57-year-old Caucasian woman diagnosed with recurrence after curative surgery for rectal adenocarcinoma stage III (ypT3N2). Despite undergoing multiple lines of standard chemotherapy, disease control could not be maintained. Consequently, regorafenib, a multikinase inhibitor with antiangiogenic proprieties, was started as a late-line treatment and a dose reduction strategy allowed a long-term response of more than 9 years with good tolerability.
Collapse
|
7
|
Bulska-Będkowska W, Czajka-Francuz P, Cisoń-Jurek S, Owczarek AJ, Francuz T, Chudek J. Predictive Role of Soluble IL-6R, TNF-R1/2, and Cell Adhesion Molecules Serum Levels in the Preoperative and Adjuvant Therapy in Women with Nonmetastatic Breast Cancer: A Preliminary Study. J Interferon Cytokine Res 2022; 42:557-567. [PMID: 36130158 DOI: 10.1089/jir.2022.0092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Soluble cell adhesion molecules (sCAMs) are involved in the development of neoplastic diseases. sCAMs can block lymphocytes and promote angiogenesis and migration of breast cancer (BC) cells. Interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) enhance metastatic potential via upregulation of CAMs. We assessed soluble interleukin-6 receptor subunit alpha (IL-6Ra), TNF-R1, TNF-R2, E-selectin, P-selectin, VCAM-1, ICAM-1, and EpCAM in 89 women with stage I-III BC and 28 healthy women. Blood samples were obtained at the beginning of neoadjuvant/induction (N = 49) or adjuvant treatment (N = 40), and after 2 months. Surgery revealed complete response in 29.4% of patients, partial response in 67%, and stable disease in 5.9%. Achieving a pathological response was 4 times greater for baseline levels of sIL-6Ra >5.63 ng/mL [odds ratio (OR) = 4.1, 95% confidence interval (CI): 0.8-20.4, P = 0.08] and more than 6 times for soluble tumor necrosis factor receptor 1 (sTNF-R1) ≥ 0.97 ng/mL (OR = 6.2, 95% CI: 1.2-32.3, P < 0.05). Compared with the control group, serum sP-selectin, soluble epithelial cell adhesion molecule (sEpCAM), and sTNF-R2 concentrations were significantly higher in patients who started adjuvant therapy (P < 0.05) and preoperative therapy (P < 0.01). Baseline serum sIL-6Ra concentrations were significantly higher in patients before surgery than in patients after tumor resection (P < 0.05), independent of the follow-up time. The baseline serum soluble receptors of IL-6 (sIL-6R) and TNF-α (sTNF-R1) concentrations have a predictive value for preoperative therapy in patients with BC.
Collapse
Affiliation(s)
- Weronika Bulska-Będkowska
- Department of Internal Diseases and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Paulina Czajka-Francuz
- Department of Internal Diseases and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Sylwia Cisoń-Jurek
- Department of Internal Diseases and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Aleksander J Owczarek
- Health Promotion and Obesity Management Unit, Department of Pathophysiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Tomasz Francuz
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Jerzy Chudek
- Department of Internal Diseases and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
8
|
Diagnostic and Prognostic Roles of Thrombospondin-2 in Digestive System Cancers. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3749306. [PMID: 35872838 PMCID: PMC9303135 DOI: 10.1155/2022/3749306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 06/27/2022] [Indexed: 12/24/2022]
Abstract
Background Cancers of digestive system have high case-fatality rate. It is important to find more appropriate methods in diagnosing and predicting gastrointestinal malignances. And thrombospondin-2 (TSP-2) was reported to have the functions, although results were not identical. So we performed this meta-analysis to clarify the significance of TSP-2 in this area. Methods PubMed, Embase, Web of Science, Cochrane Library, and Clinicaltrial.gov were searched for relevant studies. Data were extracted from these involved records. For the meta-analysis of diagnostic test, bivariate mixed effect model was used to estimate diagnostic accuracy. For prognosis part, HRs and their 95% CIs were pooled to compare the overall survival (OS) and disease-free survival (DFS) between patients with high TSP-2 and low TSP-2. Results Nine records were eligible for the analysis of diagnostic test. Pooled results were as follows: sensitivity 0.60 (0.52, 0.68), specificity 0.96 (0.91, 0.98), positive likelihood ratio (PLR) 15.4 (7.3, 32.2), negative likelihood ratio (NLR) 0.42 (0.34, 0.50), and diagnostic odds ratio (DOR) 37 (18, 76). While in prognosis part, 10 articles were included. Patients with increased TSP-2 had shorter OS (HR = 1.64, 95% CI = 1.21-2.22); however, no difference was found in DFS between TSP-2 high and low groups (HR = 1.44, 95% CI = 0.28-7.33). Conclusions TSP-2, as a diagnostic marker, has a high specificity but a moderate sensitivity. Meanwhile, it plays a role in predicting OS. Therefore, making TSP-2 a routine assay could be beneficial to high-risk individuals and patients with digestive malignances.
Collapse
|
9
|
Huang M, Lin Y, Wang C, Deng L, Chen M, Assaraf YG, Chen ZS, Ye W, Zhang D. New insights into antiangiogenic therapy resistance in cancer: Mechanisms and therapeutic aspects. Drug Resist Updat 2022; 64:100849. [PMID: 35842983 DOI: 10.1016/j.drup.2022.100849] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Angiogenesis is a hallmark of cancer and is required for tumor growth and progression. Antiangiogenic therapy has been revolutionarily developing and was approved for the treatment of various types of cancer for nearly two decades, among which bevacizumab and sorafenib continue to be the two most frequently used antiangiogenic drugs. Although antiangiogenic therapy has brought substantial survival benefits to many cancer patients, resistance to antiangiogenic drugs frequently occurs during clinical treatment, leading to poor outcomes and treatment failure. Cumulative evidence has demonstrated that the intricate interplay among tumor cells, bone marrow-derived cells, and local stromal cells critically allows for tumor escape from antiangiogenic therapy. Currently, drug resistance has become the main challenge that hinders the therapeutic efficacies of antiangiogenic therapy. In this review, we describe and summarize the cellular and molecular mechanisms conferring tumor drug resistance to antiangiogenic therapy, which was predominantly associated with redundancy in angiogenic signaling molecules (e.g., VEGFs, GM-CSF, G-CSF, and IL17), alterations in biological processes of tumor cells (e.g., tumor invasiveness and metastasis, stemness, autophagy, metabolic reprogramming, vessel co-option, and vasculogenic mimicry), increased recruitment of bone marrow-derived cells (e.g., myeloid-derived suppressive cells, tumor-associated macrophages, and tumor-associated neutrophils), and changes in the biological functions and features of local stromal cells (e.g., pericytes, cancer-associated fibroblasts, and endothelial cells). We also review potential biomarkers to predict the response to antiangiogenic therapy in cancer patients, which mainly consist of imaging biomarkers, cellular and extracellular proteins, a certain type of bone marrow-derived cells, local stromal cell content (e.g., pericyte coverage) as well as serum or plasma biomarkers (e.g., non-coding RNAs). Finally, we highlight the recent advances in combination strategies with the aim of enhancing the response to antiangiogenic therapy in cancer patients and mouse models. This review introduces a comprehensive understanding of the mechanisms and biomarkers associated with the evasion of antiangiogenic therapy in cancer, providing an outlook for developing more effective approaches to promote the therapeutic efficacy of antiangiogenic therapy.
Collapse
Affiliation(s)
- Maohua Huang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Yuning Lin
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Chenran Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Lijuan Deng
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Minfeng Chen
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Institute for Biotechnology, St. John's University, NY 11439, USA.
| | - Wencai Ye
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| | - Dongmei Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
10
|
Mori K, Schuettfort VM, Katayama S, Laukhtina E, Pradere B, Quhal F, Sari Motlagh R, Mostafaei H, Grossmann NC, Rajwa P, Teoh JY, Resch I, Fajkovic H, Moschini M, D'andrea D, Abufaraj M, Karakiewicz PI, Lotan Y, Scherr D, Egawa S, Compérat E, Shariat SF. Prognostic Role of Preoperative Vascular Cell Adhesion Molecule-1 Plasma Levels in Urothelial Carcinoma of the Bladder Treated With Radical Cystectomy. Ann Surg Oncol 2022; 29:5307-5316. [PMID: 35347517 PMCID: PMC9246812 DOI: 10.1245/s10434-022-11575-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 02/21/2022] [Indexed: 01/19/2023]
Abstract
Abstract
Background
Angiogenesis-related marker vascular cell adhesion molecule-1 (VCAM-1) has been shown to be elevated in urothelial carcinoma of the bladder (UCB), but its predictive/prognostic role has not been determined. Thus, this study aimed to investigate the predictive/prognostic role of VCAM-1 for patients who have UCB treated with radical cystectomy (RC).
Methods
The study enrolled 1036 patients with clinically non-metastatic advanced UCB who underwent RC, and plasma VCAM-1 was evaluated preoperatively. The correlation of plasma VCAM-1 with pathologic and survival outcomes was assessed using binominal logistic regression and multivariable Cox regression analyses. Discrimination was assessed using the area under the curve and concordance indices. The clinical net benefit was evaluated using decision curve analysis (DCA).
Results
Preoperative VCAM-1 was significantly elevated in patients with adverse pathologic features. Higher VCAM-1 levels were independently associated with increased risk of lymph-node-metastasis (LNM), ≥pT3 disease, and non-organ-confined disease (NOCD (p < 0.001 for each). Preoperative plasma VCAM-1 was independently associated with recurrence-free survival (RFS), cancer-specific survival (CSS), and overall survival (OS) in pre- and postoperative multivariable models. Adding VCAM-1 to these predictive models improved their discriminatory ability to predict all outcomes by a significant margin. In the DCA, VCAM-1 addition to the reference models for prediction of LNM, NOCD, RFS, and CSS resulted in relevant improvement.
Conclusions
Elevated plasma VCAM-1 was associated with biologically and clinically aggressive UCB disease features. After validation, preoperative VCAM-1 may serve as a biomarker to help identify patients likely to benefit from intensified/multimodal therapy. In addition, VCAM-1 improved the discriminatory power of predictive/prognostic models and can be used to refine personalized clinical decision-making.
Collapse
Affiliation(s)
- Keiichiro Mori
- Department of Urology, Medical University of Vienna, Vienna, Austria
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Victor M Schuettfort
- Department of Urology, Medical University of Vienna, Vienna, Austria
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Satoshi Katayama
- Department of Urology, Medical University of Vienna, Vienna, Austria
- Department of Urology, Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Ekaterina Laukhtina
- Department of Urology, Medical University of Vienna, Vienna, Austria
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
| | - Benjamin Pradere
- Department of Urology, Medical University of Vienna, Vienna, Austria
- Department of Urology, CHRU Tours, Tours, France
- Université François Rabelais de Tours, PRES Centre Val de Loire, Tours, France
| | - Fahad Quhal
- Department of Urology, Medical University of Vienna, Vienna, Austria
- Department of Urology, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Reza Sari Motlagh
- Department of Urology, Medical University of Vienna, Vienna, Austria
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hadi Mostafaei
- Department of Urology, Medical University of Vienna, Vienna, Austria
- Research Center for Evidence Based Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nico C Grossmann
- Department of Urology, Medical University of Vienna, Vienna, Austria
- Department of Urology, University Hospital Zurich, Zurich, Switzerland
| | - Pawel Rajwa
- Department of Urology, Medical University of Vienna, Vienna, Austria
- Department of Urology, Medical University of Silesia, Zabrze, Poland
| | - Jeremy Yc Teoh
- Department of Surgery, S.H. Ho Urology Centre, The Chinese University of Hong Kong, Hong Kong, China
| | - Irene Resch
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Harun Fajkovic
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Marco Moschini
- Department of Urology, Medical University of Vienna, Vienna, Austria
- Klinik für Urologie, Luzerner Kantonsspital, Lucerne, Switzerland
| | - David D'andrea
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Mohammad Abufaraj
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Pierre I Karakiewicz
- Cancer Prognostics and Health Outcomes Unit, University of Montreal Health Centre, Montreal, Canada
| | - Yair Lotan
- Department of Urology, University of Texas Southwestern, Dallas, TX, USA
| | - Douglas Scherr
- Department of Urology, Weill Cornell Medical College, New York, NY, USA
| | - Shin Egawa
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Eva Compérat
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Shahrokh F Shariat
- Department of Urology, Medical University of Vienna, Vienna, Austria.
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia.
- Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan.
- Department of Urology, University of Texas Southwestern, Dallas, TX, USA.
- Department of Urology, Weill Cornell Medical College, New York, NY, USA.
- Department of Urology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.
- Karl Landsteiner Institute of Urology and Andrology, Vienna, Austria.
| |
Collapse
|
11
|
Amlodipine improves the outcomes of regorafenib in metastatic colorectal cancer. Anticancer Drugs 2022; 33:389-393. [DOI: 10.1097/cad.0000000000001273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
12
|
Jia J, Howard L, Liu Y, Starr MD, Brady JC, Niedzwiecki D, Strickler JH, Nixon AB. Cabozantinib with or without Panitumumab for RAS wild-type metastatic colorectal cancer: impact of MET amplification on clinical outcomes and circulating biomarkers. Cancer Chemother Pharmacol 2022; 89:413-422. [PMID: 35171350 DOI: 10.1007/s00280-022-04404-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/29/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE Acquired resistance to EGFR inhibitors in metastatic colorectal cancer (mCRC) remains a hurdle for effective treatment. MET amplification has been indicated as a driver of acquired resistance. Clinical activity has been demonstrated for the combination of EGFR and MET inhibitors in mCRC. But the impact of this regimen on angiogenesis and inflammation remains largely unknown. METHODS In this non-randomized, open-label phase Ib/II study, four patients were treated with cabozantinib alone and 25 patients received the combination of cabozantinib and panitumumab. MET amplification was detected in blood in all four patients treated with cabozantinib monotherapy and 5/25 patients treated with cabozantinib and panitumumab combination therapy. Plasma samples from 28 patients were available for biomarker analysis. RESULTS A panel of circulating protein biomarkers was assessed in patient plasma at baseline and on-treatment. Baseline marker levels were analyzed for prognostic value for clinical outcomes, including MET amplification as a covariate. HGF and OPN were prognostic for both progression-free survival (PFS) and overall survival (OS), while six markers (IL-6, VCAM-1, VEGF-R1, TSP-2, TIMP-1, ICAM-1) were prognostic only for OS. In patients with MET amplification, baseline PDGF-AA, PDGF-BB, TGF-β1, and VEGF-C levels were significantly higher, whereas baseline TGFβ-R3 levels were significantly lower than MET non-amplified patients. On-treatment change of four markers (CD73, PlGF, PDGF-BB, VEGF) were significantly different between MET amplified and non-amplified subpopulations. CONCLUSION This study identified circulating HGF and several inflammatory and angiogenic proteins as prognostic biomarkers. Furthermore, MET amplification status is associated with both baseline expression and on-treatment modulation of members of angiogenesis and TGF-β pathway proteins. CLINICAL TRIALS REGISTRATION NUMBER ClinicalTrials.gov identifier: NCT02008383.
Collapse
Affiliation(s)
- Jingquan Jia
- Duke Cancer Institute, Durham, NC, USA.,Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Lauren Howard
- Duke Department of Biostatistics and Bioinformatics, Durham, NC, USA
| | - Yingmiao Liu
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Mark D Starr
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - John C Brady
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Donna Niedzwiecki
- Duke Department of Biostatistics and Bioinformatics, Durham, NC, USA
| | - John H Strickler
- Duke Cancer Institute, Durham, NC, USA.,Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Andrew B Nixon
- Duke Cancer Institute, Durham, NC, USA. .,Department of Medicine, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
13
|
Wetzel-Strong SE, Weinsheimer S, Nelson J, Pawlikowska L, Clark D, Starr MD, Liu Y, Kim H, Faughnan ME, Nixon AB, Marchuk DA. Pilot investigation of circulating angiogenic and inflammatory biomarkers associated with vascular malformations. Orphanet J Rare Dis 2021; 16:372. [PMID: 34479577 PMCID: PMC8414780 DOI: 10.1186/s13023-021-02009-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/24/2021] [Indexed: 01/03/2023] Open
Abstract
Background Vascular malformations in the central nervous system are difficult to monitor and treat due to their inaccessible location. Angiogenic and inflammatory proteins are secreted into the bloodstream and may serve as useful biomarkers for identifying patients at risk for complications or with certain disease phenotypes.
Methods A validated multiplex protein array consisting of 26 angiogenic and inflammatory biomarkers (Angiome) was assessed in plasma isolated from healthy controls and patients with either sporadic brain arteriovenous malformation (BAVM), familial cerebral cavernous malformation (CCM), or hereditary hemorrhagic telangiectasia (HHT). These samples were obtained from archives of ongoing research studies at the University of California San Francisco and through prospective collection at the Toronto HHT Centre at St. Michael’s Hospital. Results We compared circulating biomarker levels from each patient group to healthy controls and analyzed each pairwise combination of patient groups for differences in biomarker levels. Additionally, we analyzed the HHT samples to determine the association between biomarker levels and the following HHT-specific phenotypes, BAVM, pulmonary arteriovenous malformation (PAVM), liver vascular malformation (LVM), and gastrointestinal (GI) bleeding. Compared to controls, levels of SDF1 were significantly elevated in HHT patients (Proportional Increase [PI] = 1.87, p < 0.001, q = 0.011). Levels of sENG were significantly reduced in HHT patients compared to controls (PI = 0.56, p < 0.001, q < 0.001), reflecting the prevalence of HHT1 patients in this cohort. Levels of IL6 (PI = 3.22, p < 0.001, q < 0.001) and sTGFβR3 (PI = 0.70, p = 0.001, q < 0.029) differed significantly in CCM patients compared to controls. Compared to controls, ten of the biomarkers were significantly different in sporadic BAVM patients (q-values < 0.05). Among the pairwise combinations of patient groups, a significant elevation was observed in TGFβ1 in CCM patients compared to sporadic BAVM patients (PI = 2.30, p < 0.001, q = 0.034). When examining the association of circulating biomarker levels with HHT-specific phenotypes, four markers were significantly lower in HHT patients with BAVM (q-values < 0.05), and four markers were significantly higher in patients with LVM (q-values < 0.05). Conclusions This pilot study suggests that the profile of circulating angiogenic and inflammatory biomarkers may be unique to each type of vascular malformation. Furthermore, this study indicates that circulating biomarkers may be useful for assessing phenotypic traits of vascular malformations. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-02009-7.
Collapse
Affiliation(s)
- Sarah E Wetzel-Strong
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 265 CARL Bldg., Box #3175 DUMC, Durham, NC, 27710, USA
| | - Shantel Weinsheimer
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey Nelson
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Ludmila Pawlikowska
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Dewi Clark
- Toronto HHT Centre, St. Michael's Hospital and the Li Ka Shing Knowledge Institute, Toronto, Canada
| | - Mark D Starr
- Department of Medicine, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Yingmiao Liu
- Department of Medicine, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Helen Kim
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Marie E Faughnan
- Toronto HHT Centre, St. Michael's Hospital and the Li Ka Shing Knowledge Institute, Toronto, Canada.,Division of Respirology, Department of Medicine, University of Toronto, Toronto, Canada
| | - Andrew B Nixon
- Department of Medicine, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Douglas A Marchuk
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 265 CARL Bldg., Box #3175 DUMC, Durham, NC, 27710, USA.
| |
Collapse
|
14
|
Armstrong AJ, Nixon AB, Carmack A, Yang Q, Eisen T, Stadler WM, Jones RJ, Garcia JA, Vaishampayan UN, Picus J, Hawkins RE, Hainsworth JD, Kollmannsberger CK, Logan TF, Puzanov I, Pickering LM, Ryan CW, Protheroe A, George DJ, Halabi S. Angiokines Associated with Targeted Therapy Outcomes in Patients with Non-Clear Cell Renal Cell Carcinoma. Clin Cancer Res 2021; 27:3317-3328. [PMID: 33593885 DOI: 10.1158/1078-0432.ccr-20-4504] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/11/2021] [Accepted: 02/11/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Biomarkers are needed in patients with non-clear cell renal cell carcinomas (NC-RCC) to inform treatment selection but also to identify novel therapeutic targets. We thus sought to profile circulating angiokines in the context of a randomized treatment trial of everolimus versus sunitinib. PATIENTS AND METHODS ASPEN (NCT01108445) was an international, randomized, open-label phase II trial of patients with metastatic papillary, chromophobe, or unclassified NC-RCC with no prior systemic therapy. Patients were randomized to everolimus or sunitinib and treated until disease progression or unacceptable toxicity. The primary endpoint was radiographic progression-free survival (PFS) defined by RECIST 1.1. Plasma angiokines were collected at baseline, cycle 3, and progression and associated with PFS and overall survival (OS). RESULTS We enrolled 108 patients, 51 received sunitinib and 57 everolimus; of these, 99 patients had evaluable plasma for 23 angiokines. At the final data cutoff, 94 PFS and 64 mortality events had occurred. Angiokines that were independently adversely prognostic for OS were osteopontin (OPN), TIMP-1, thrombospondin-2 (TSP-2), hepatocyte growth factor (HGF), and VCAM-1, and these were also associated with poor-risk disease. Stromal derived factor 1 (SDF-1) was associated with improved survival. OPN was also significantly associated with worse PFS. No statistically significant angiokine-treatment outcome interactions were observed for sunitinib or everolimus. Angiopoeitin-2 (Ang-2), CD-73, HER-3, HGF, IL6, OPN, PIGF, PDGF-AA, PDGF-BB, SDF-1, TGF-b1-b2, TGFb-R3, TIMP-1, TSP-2, VCAM-1, VEGF, and VEGF-R1 levels increased with progression on everolimus, while CD-73, ICAM-1, IL6, OPN, PlGF, SDF-1, TGF-b2, TGFb-R3, TIMP-1, TSP-2, VEGF, VEGF-D, and VCAM-1 increased with progression on sunitinib. CONCLUSIONS In patients with metastatic NC-RCC, we identified several poor prognosis angiokines and immunomodulatory chemokines during treatment with sunitinib or everolimus, particularly OPN.
Collapse
Affiliation(s)
- Andrew J Armstrong
- Duke University Department of Medicine, Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, North Carolina.
| | - Andrew B Nixon
- Duke University Department of Medicine, Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, North Carolina
| | - Andrea Carmack
- Duke University Department of Medicine, Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, North Carolina
| | - Qian Yang
- Duke University Department of Medicine, Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, North Carolina
| | - Tim Eisen
- University of Cambridge, Cambridge, United Kingdom
| | | | - Robert J Jones
- University of Glasgow, The Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | | | - Ulka N Vaishampayan
- University of Michigan/Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Joel Picus
- Washington University in St. Louis, St. Louis, Missouri
| | | | | | | | - Theodore F Logan
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, Indiana
| | - Igor Puzanov
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | | | - Christopher W Ryan
- Oregon Health & Science University, OHSU Knight Cancer Institute, Portland, Oregon
| | - Andrew Protheroe
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Daniel J George
- Duke University Department of Medicine, Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, North Carolina
| | - Susan Halabi
- Department of Biostatistics, Duke University, Durham, North Carolina
| |
Collapse
|
15
|
Nakajima H, Fukuoka S, Masuishi T, Takashima A, Kumekawa Y, Kajiwara T, Yamazaki K, Negoro Y, Komoda M, Makiyama A, Denda T, Hatachi Y, Suto T, Sugimoto N, Enomoto M, Ishikawa T, Kashiwada T, Ando K, Yuki S, Okuyama H, Kusaba H, Sakai D, Okamoto K, Tamura T, Yamashita K, Gosho M, Moriwaki T. Clinical Impact of Primary Tumor Location in Metastatic Colorectal Cancer Patients Under Later-Line Regorafenib or Trifluridine/Tipiracil Treatment. Front Oncol 2021; 11:688709. [PMID: 34211856 PMCID: PMC8239287 DOI: 10.3389/fonc.2021.688709] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 04/30/2021] [Indexed: 12/27/2022] Open
Abstract
Background Primary tumor location (PTL) is an important prognostic and predictive factor in the first-line treatment of metastatic colorectal cancer (mCRC). Although regorafenib (REG) and trifluridine/tipiracil (FTD/TPI) have been introduced recently, the clinical impact of PTL in these treatments is not well understood. Materials and Methods We retrospectively evaluated patients with mCRC who were registered in a multicenter observational study (the REGOTAS study). The main inclusion criteria were Eastern Cooperative Oncology Group performance status (ECOG PS) of 0-2, refractory or intolerant to fluoropyrimidines, oxaliplatin, irinotecan, angiogenesis inhibitors, anti-epidermal growth factor receptor therapy (if RAS wild-type), and no prior use of REG and FTD/TPI. The impact of PTL on overall survival (OS) was evaluated using Cox proportional hazard models based on baseline characteristics. Results A total of 550 patients (223 patients in the REG group and 327 patients in the FTD/TPI group) were included in this study, with 122 patients with right-sided tumors and 428 patients with left-sided tumors. Although the right-sided patients had significantly shorter OS compared with the left-sided patients by univariate analysis (p = 0.041), a multivariate analysis revealed that PTL was not an independent prognostic factor (hazard ratio, 0.95; p = 0.64). In a subgroup analysis, the OS was comparable between the REG and FTD/TPI groups regardless of PTL (p for interactions = 0.60). Conclusions In the present study, PTL is not a prognostic and predictive factor in patients with mCRC under later-line REG or FTD/TPI therapy.
Collapse
Affiliation(s)
- Hiromichi Nakajima
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Shota Fukuoka
- Division of Cancer Immunology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Toshiki Masuishi
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Aichi, Japan
| | - Atsuo Takashima
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Yosuke Kumekawa
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Takeshi Kajiwara
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Ehime, Japan
| | - Kentaro Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Yuji Negoro
- Clinical Oncology Division, Kochi Health Sciences Center, Kochi, Japan
| | - Masato Komoda
- Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Akitaka Makiyama
- Department of Hematology/Oncology, Japan Community Healthcare Organization Kyushu Hospital, Fukuoka, Japan.,Cancer Center, Gifu University Hospital, Gifu, Japan
| | - Tadamichi Denda
- Division of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Yukimasa Hatachi
- Department of Clinical Oncology, Kansai Rosai Hospital, Hyogo, Japan
| | - Takeshi Suto
- Department of Gastroenterological Surgery, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Naotoshi Sugimoto
- Department of Medical Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Masanobu Enomoto
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, Tokyo, Japan
| | - Toshiaki Ishikawa
- Department of Specialized Surgeries, Graduate School of Medicine and Dentistry, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomomi Kashiwada
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Koji Ando
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Satoshi Yuki
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Hokkaido, Japan
| | - Hiroyuki Okuyama
- Department of Clinical Oncology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Hitoshi Kusaba
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daisuke Sakai
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichi Okamoto
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Takao Tamura
- Department of Medical Oncology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Kimihiro Yamashita
- Division of Gastrointestinal Surgery, Department of Surgery, Graduate School of Medicine, Kobe University, Hyogo, Japan
| | - Masahiko Gosho
- Department of Biostatistics, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Toshikazu Moriwaki
- Department of Gastroenterology, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
16
|
Avolio M, Trusolino L. Rational Treatment of Metastatic Colorectal Cancer: A Reverse Tale of Men, Mice, and Culture Dishes. Cancer Discov 2021; 11:1644-1660. [PMID: 33820776 DOI: 10.1158/2159-8290.cd-20-1531] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/04/2021] [Accepted: 02/01/2021] [Indexed: 11/16/2022]
Abstract
Stratification of colorectal cancer into subgroups with different response to therapy was initially guided by descriptive associations between specific biomarkers and treatment outcome. Recently, preclinical models based on propagatable patient-derived tumor samples have yielded an improved understanding of disease biology, which has facilitated the functional validation of correlative information and the discovery of novel response determinants, therapeutic targets, and mechanisms of tumor adaptation and drug resistance. We review the contribution of patient-derived models to advancing colorectal cancer characterization, discuss their influence on clinical decision-making, and highlight emerging challenges in the interpretation and clinical transferability of results obtainable with such approaches. SIGNIFICANCE: Association studies in patients with colorectal cancer have led to the identification of response biomarkers, some of which have been implemented as companion diagnostics for therapeutic decisions. By enabling biological investigation in a clinically relevant experimental context, patient-derived colorectal cancer models have proved useful to examine the causal role of such biomarkers in dictating drug sensitivity and are providing fresh knowledge on new actionable targets, dynamics of tumor evolution and adaptation, and mechanisms of drug resistance.
Collapse
Affiliation(s)
- Marco Avolio
- Department of Oncology, University of Torino, Candiolo, Torino, Italy.,Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
| | - Livio Trusolino
- Department of Oncology, University of Torino, Candiolo, Torino, Italy. .,Candiolo Cancer Institute - FPO IRCCS, Candiolo, Torino, Italy
| |
Collapse
|
17
|
Liu Y, Paauwe M, Nixon AB, Hawinkels LJ. Endoglin Targeting: Lessons Learned and Questions That Remain. Int J Mol Sci 2020; 22:ijms22010147. [PMID: 33375670 PMCID: PMC7795616 DOI: 10.3390/ijms22010147] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
Approximately 30 years ago, endoglin was identified as a transforming growth factor (TGF)-β coreceptor with a crucial role in developmental biology and tumor angiogenesis. Its selectively high expression on tumor vessels and its correlation with poor survival in cancer patients led to the exploration of endoglin as a therapeutic target for cancer. The endoglin neutralizing antibody TRC105 (Carotuximab®, Tracon Pharmaceuticals (San Diego, CA, USA) was subsequently tested in a wide variety of preclinical cancer models before being tested in phase I-III clinical studies in cancer patients as both a monotherapy and in combination with other chemotherapeutic and anti-angiogenic therapies. The combined data of these studies have revealed new insights into the role of endoglin in angiogenesis and its expression and functional role on other cells in the tumor microenvironment. In this review, we will summarize the preclinical work, clinical trials and biomarker studies of TRC105 and explore what these studies have enabled us to learn and what questions remain unanswered.
Collapse
Affiliation(s)
- Yingmiao Liu
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; (Y.L.); (A.B.N.)
| | - Madelon Paauwe
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Andrew B. Nixon
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA; (Y.L.); (A.B.N.)
| | - Lukas J.A.C. Hawinkels
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
- Correspondence: ; Tel.: +31-71-526-6736
| |
Collapse
|