151
|
Zaman S, Jadid H, Denson AC, Gray JE. Targeting Trop-2 in solid tumors: future prospects. Onco Targets Ther 2019; 12:1781-1790. [PMID: 30881031 PMCID: PMC6402435 DOI: 10.2147/ott.s162447] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Trop-2 is a transmembrane glycoprotein that is upregulated in all cancer types independent of baseline levels of Trop-2 expression. Trop-2 is an ideal candidate for targeted therapeutics due to it being a transmembrane protein with an extracellular domain overexpressed on a wide variety of tumors as well as its upregulated expression relative to normal cells. As a result, several Trop-2-targeted therapeutics have recently been developed for clinical use, such as anti-Trop-2 antibodies and Trop-2-targeted antibody-drug conjugates (ADC). Subsequently, multiple early-phase clinical trials have demonstrated safety and clinical benefit of Trop-2-based ADCs across multiple tumor types. This includes clinical benefit and tolerability in tumor types with limited treatment options, such as triple-negative breast cancer, platinum-resistant urothelial cancer, and small-cell lung cancer. In this review, we elaborate on all clinical trials involving Trop-2.
Collapse
Affiliation(s)
- Saif Zaman
- Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Hassan Jadid
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA,
| | - Aaron C Denson
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA,
| | - Jhanelle E Gray
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA,
| |
Collapse
|
152
|
Bardia A, Mayer IA, Vahdat LT, Tolaney SM, Isakoff SJ, Diamond JR, O'Shaughnessy J, Moroose RL, Santin AD, Abramson VG, Shah NC, Rugo HS, Goldenberg DM, Sweidan AM, Iannone R, Washkowitz S, Sharkey RM, Wegener WA, Kalinsky K. Sacituzumab Govitecan-hziy in Refractory Metastatic Triple-Negative Breast Cancer. N Engl J Med 2019; 380:741-751. [PMID: 30786188 DOI: 10.1056/nejmoa1814213] [Citation(s) in RCA: 564] [Impact Index Per Article: 94.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Standard chemotherapy is associated with low response rates and short progression-free survival among patients with pretreated metastatic triple-negative breast cancer. Sacituzumab govitecan-hziy is an antibody-drug conjugate that combines a humanized monoclonal antibody, which targets the human trophoblast cell-surface antigen 2 (Trop-2), with SN-38, which is conjugated to the antibody by a cleavable linker. Sacituzumab govitecan-hziy enables delivery of high concentrations of SN-38 to tumors. METHODS We conducted a phase 1/2 single-group, multicenter trial involving patients with advanced epithelial cancers who received sacituzumab govitecan-hziy intravenously on days 1 and 8 of each 21-day cycle until disease progression or unacceptable toxic effects. A total of 108 patients received sacituzumab govitecan-hziy at a dose of 10 mg per kilogram of body weight after receiving at least two previous anticancer therapies for metastatic triple-negative breast cancer. The end points included safety; the objective response rate (according to Response Evaluation Criteria in Solid Tumors, version 1.1), which was assessed locally; the duration of response; the clinical benefit rate (defined as a complete or partial response or stable disease for at least 6 months); progression-free survival; and overall survival. Post hoc analyses determined the response rate and duration, which were assessed by blinded independent central review. RESULTS The 108 patients with triple-negative breast cancer had received a median of 3 previous therapies (range, 2 to 10). Four deaths occurred during treatment; 3 patients (2.8%) discontinued treatment because of adverse events. Grade 3 or 4 adverse events (in ≥10% of the patients) included anemia and neutropenia; 10 patients (9.3%) had febrile neutropenia. The response rate (3 complete and 33 partial responses) was 33.3% (95% confidence interval [CI], 24.6 to 43.1), and the median duration of response was 7.7 months (95% CI, 4.9 to 10.8); as assessed by independent central review, these values were 34.3% and 9.1 months, respectively. The clinical benefit rate was 45.4%. Median progression-free survival was 5.5 months (95% CI, 4.1 to 6.3), and overall survival was 13.0 months (95% CI, 11.2 to 13.7). CONCLUSIONS Sacituzumab govitecan-hziy was associated with durable objective responses in patients with heavily pretreated metastatic triple-negative breast cancer. Myelotoxic effects were the main adverse reactions. (Funded by Immunomedics; IMMU-132-01 ClinicalTrials.gov number, NCT01631552.).
Collapse
Affiliation(s)
- Aditya Bardia
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - Ingrid A Mayer
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - Linda T Vahdat
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - Sara M Tolaney
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - Steven J Isakoff
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - Jennifer R Diamond
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - Joyce O'Shaughnessy
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - Rebecca L Moroose
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - Alessandro D Santin
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - Vandana G Abramson
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - Nikita C Shah
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - Hope S Rugo
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - David M Goldenberg
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - Ala M Sweidan
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - Robert Iannone
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - Sarah Washkowitz
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - Robert M Sharkey
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - William A Wegener
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| | - Kevin Kalinsky
- From the Massachusetts General Hospital Cancer Center (A.B., S.J.I.) and Dana-Farber Cancer Institute (S.M.T.), Harvard Medical School, Boston; Vanderbilt-Ingram Cancer Center, Nashville (I.A.M., V.G.A.); Weill Cornell Medical College (L.T.V.) and New York-Presbyterian-Columbia University Irving Medical Center (K.K.), New York; University of Colorado Cancer Center, Aurora (J.R.D.); Texas Oncology, Baylor University Medical Center, US Oncology, Dallas (J.O.); Orlando Health University of Florida Health Cancer Center, Orlando (R.L.M., N.C.S.); Yale University School of Medicine, New Haven, CT (A.D.S.); University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco (H.S.R.); Immunomedics, Morris Plains, NJ (D.M.G., R.I., S.W., R.M.S., W.A.W.); and AIS Consulting, Ann Arbor, MI (A.M.S.)
| |
Collapse
|
153
|
Zhang J, Ma H, Yang L, Yang H, He Z. Silencing of the TROP2 gene suppresses proliferation and invasion of hepatocellular carcinoma HepG2 cells. J Int Med Res 2019; 47:1319-1329. [PMID: 30678511 PMCID: PMC6421384 DOI: 10.1177/0300060518822913] [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] [Indexed: 01/07/2023] Open
Abstract
Objectives Overexpression of human trophoblast cell surface antigen 2 (Trop2) has been
observed in many cancers; however, its roles in proliferation, apoptosis,
migration, and invasion of hepatocellular carcinoma (HCC) remain unclear.
Thus, this study aimed to characterize the function of Trop2 in HCC. Methods Trop2 protein expression was detected by immunohistochemistry in HCC tissues.
Cell proliferation, apoptosis, and invasion were respectively measured by
CCK-8, flow cytometry, Transwell, and wound healing assays. Expression
levels of epithelial–mesenchymal transition-related proteins and Trop2
protein in HCC cell lines were detected by western blotting after silencing
of the TROP2 gene. Results Trop2 protein was highly expressed in HCC tissues and HCC cell lines. Trop2
mRNA and protein expression levels decreased in HepG2 and HCCLM3 cells after
transfection with Trop2 siRNA. Silencing of the TROP2 gene
in HepG2 and HCCLM3 cells strongly inhibited cell proliferation and
migration, while enhancing cell apoptosis. Investigation of the molecular
mechanism revealed that silencing of the TROP2 gene
suppressed epithelial–mesenchymal transition of HepG2 and HCCLM3 cells. Conclusions The results of the present study may improve understanding of the role of
Trop2 in regulation of cell proliferation and invasion, and may aid in
development of novel therapy for HCC.
Collapse
Affiliation(s)
- Jian Zhang
- Department of General Surgery, Central Hospital of Nanchong, Nanchong, Sichuan Province, China
| | - Hai Ma
- Department of General Surgery, Central Hospital of Nanchong, Nanchong, Sichuan Province, China
| | - Liu Yang
- Department of respiratory medicine, Central Hospital of Nanchong, Nanchong, Sichuan Province, China
| | - Hongchun Yang
- Department of General Surgery, Central Hospital of Nanchong, Nanchong, Sichuan Province, China
| | - Zhenxing He
- Department of General Surgery, Central Hospital of Nanchong, Nanchong, Sichuan Province, China
| |
Collapse
|
154
|
Abdou AG, Shabaan M, Abdallha R, Nabil N. Diagnostic Value of TROP-2 and CK19 Expression in Papillary Thyroid Carcinoma in Both Surgical and Cytological Specimens. CLINICAL PATHOLOGY (THOUSAND OAKS, VENTURA COUNTY, CALIF.) 2019; 12:2632010X19863047. [PMID: 31384753 PMCID: PMC6647210 DOI: 10.1177/2632010x19863047] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 06/18/2019] [Indexed: 12/29/2022]
Abstract
Papillary thyroid carcinoma (PTC) represents the most common primary malignant thyroid tumor and its diagnosis is dependent on the presence of classic nuclear features that are sometimes seen in some non-neoplastic and benign lesions. Several immunohistochemical markers are used individually or in combination to help in differentiation of PTC from mimickers. The aim of the current study was to assess the diagnostic value of TROP-2 and cytokeratin 19 (CK19) expression in differentiating PTC from other mimickers both singly and in combination. The current study was carried out on 77 surgical specimens (56 PTC and 21 non-neoplastic cases) and 12 cytological specimens (4 THY2, 6 THY4, and 2 THY5). TROP-2 was negative in 81% of non-neoplastic surgical specimens and in 100% of THY2 cytological specimens while it was positive in 71.4% of PTC surgical specimens and 100% of THY4/THY5 cytological specimens. Sensitivity and specificity of TROP-2 positive expression for diagnosis of PTC in surgical specimens reached 71% and 81%, respectively, while it reached 100% for both in cytological specimens. Cytokeratin 19 showed positive expression in 85.7% of non-neoplastic surgical specimens and in 92.9% of PTC surgical specimens. Cytokeratin 19 showed negative expression in 75% of Thy2 cases while it was positive in all studied Thy4 and Thy5 cases. Sensitivity and specificity of CK19 total estimated score for diagnosis of PTC in surgical specimens were 78.6% and 66.7%, respectively, while it reached 100% and 75% in cytological specimens. Positive TROP-2 and CK19 expression in PTC were associated with lymph node metastasis. TROP-2 is a specific rather than sensitive marker while CK19 is a sensitive rather than specific marker in differentiating PTC from other mimickers in surgical specimens. The diagnostic validity of both markers was superior in diagnosis of classic PTC compared with follicular variant PTC. TROP-2 is superior to CK19 in diagnosis of PTC in cytological specimens. Both TROP-2 and CK19 could be used preoperatively in adjunct to hematoxylin and eosin for more confident diagnosis of thyroid cytology and along with radiology as predictors of lymph node metastasis.
Collapse
Affiliation(s)
- Asmaa Gaber Abdou
- Department of Pathology, Faculty of Medicine, Menoufia University, Shebein Elkom, Egypt
| | - Mohammed Shabaan
- Department of Pathology, Faculty of Medicine, Menoufia University, Shebein Elkom, Egypt
| | - Rania Abdallha
- Department of Pathology, Faculty of Medicine, Menoufia University, Shebein Elkom, Egypt
| | - Nehal Nabil
- Department of Pathology, Faculty of Medicine, Menoufia University, Shebein Elkom, Egypt
| |
Collapse
|
155
|
Relli V, Trerotola M, Guerra E, Alberti S. Distinct lung cancer subtypes associate to distinct drivers of tumor progression. Oncotarget 2018; 9:35528-35540. [PMID: 30473748 PMCID: PMC6238974 DOI: 10.18632/oncotarget.26217] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 09/26/2018] [Indexed: 12/12/2022] Open
Abstract
The main non–small-cell lung cancer (NSCLC) histopathological subtypes are lung adenocarcinomas (LUAD) and lung squamous cell carcinomas (LUSC). To identify candidate progression determinants of NSCLC subtypes, we explored the transcriptomic signatures of LUAD versus LUSC. We then investigated the prognostic impact of the identified tumor-associated determinants. This was done utilizing DNA microarray data from 2,437 NSCLC patients. An independent analysis of a case series of 994 NSCLC was conducted by next-generation sequencing, together with gene expression profiling from GEO (https://www.ncbi.nlm.nih.gov/geo/). This work led us to identify 69 distinct tumor prognostic determinants, which impact on LUAD or LUSC clinical outcome. These included key drivers of tumor growth and cell cycle, transcription factors and metabolic determinants. Such disease determinants appeared vastly different in LUAD versus LUSC, and often had opposite impact on clinical outcome. These findings indicate that distinct tumor progression pathways are at work in the two NSCLC subtypes. Notably, most prognostic determinants would go inappropriately assessed or even undetected when globally investigating unselected NSCLC. Hence, differential consideration for NSCLC subtypes should be taken into account in current clinical evaluation procedures for lung cancer.
Collapse
Affiliation(s)
- Valeria Relli
- Unit of Cancer Pathology, CeSI-MeT, University "G. d'Annunzio", Chieti, Italy
| | - Marco Trerotola
- Unit of Cancer Pathology, CeSI-MeT, University "G. d'Annunzio", Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Emanuela Guerra
- Unit of Cancer Pathology, CeSI-MeT, University "G. d'Annunzio", Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti, Italy
| | - Saverio Alberti
- Unit of Cancer Pathology, CeSI-MeT, University "G. d'Annunzio", Chieti, Italy.,Department of Biomedical Sciences, Dentistry, Morphological and Functional Imaging, University of Messina, Messina, Italy
| |
Collapse
|
156
|
Tray N, Adams S, Esteva FJ. Antibody-drug conjugates in triple negative breast cancer. Future Oncol 2018; 14:2651-2661. [PMID: 30175620 DOI: 10.2217/fon-2018-0131] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a heterogeneous disease that comprises 15-20% of all breast cancers and is more frequently seen in younger women, African-Americans, and BRCA1 expression. Advanced TNBC carries aggressive features and is associated with overall poor outcomes. Unfortunately, there are no targeted therapies available for non-BRCA associated TNBC, which remains a high unmet therapeutic need. One emerging treatment modality includes antibody-drug conjugates which are highly selective monoclonal antibodies conjugated to cytotoxic agents, designed to deliver cytotoxic drugs to antigen-expressing tumor cells. This review will highlight three antibody-drug conjugates currently being evaluated in TNBC (CDX-011, SGN-LIV1a, IMMU-132), including one that has been given Breakthrough Therapy designation from the US FDA.
Collapse
Affiliation(s)
- Nancy Tray
- Perlmutter Cancer Center at New York University Langone Health, New York, NY 10016, USA
| | - Sylvia Adams
- Perlmutter Cancer Center at New York University Langone Health, New York, NY 10016, USA
| | - Francisco J Esteva
- Perlmutter Cancer Center at New York University Langone Health, New York, NY 10016, USA
| |
Collapse
|
157
|
Matsui S, Harada K, Miyata N, Okochi H, Miyajima A, Tanaka M. Characterization of Peribiliary Gland–Constituting Cells Based on Differential Expression of Trophoblast Cell Surface Protein 2 in Biliary Tract. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2059-2073. [DOI: 10.1016/j.ajpath.2018.05.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/02/2018] [Accepted: 05/15/2018] [Indexed: 12/18/2022]
|
158
|
Vlachostergios PJ, Jakubowski CD, Niaz MJ, Lee A, Thomas C, Hackett AL, Patel P, Rashid N, Tagawa ST. Antibody-Drug Conjugates in Bladder Cancer. Bladder Cancer 2018; 4:247-259. [PMID: 30112436 PMCID: PMC6087439 DOI: 10.3233/blc-180169] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Urothelial carcinoma (UC) is characterized by expression of a plethora of cell surface antigens, thus offering opportunities for specific therapeutic targeting with use of antibody-drug conjugates (ADCs). ADCs are structured from two major constituents, a monoclonal antibody (mAb) against a specific target and a cytotoxic drug connected via a linker molecule. Several ADCs are developed against different UC surface markers, but the ones at most advanced stages of development include sacituzumab govitecan (IMMU-132), enfortumab vedotin (ASG-22CE/ASG-22ME), ASG-15ME for advanced UC, and oportuzumab monatox (VB4-845) for early UC. Several new targets are identified and utilized for novel or existing ADC testing. The most promising ones include human epidermal growth factor receptor 2 (HER2) and members of the fibroblast growth factor receptor axis (FGF/FGFR). Positive preclinical and early clinical results are reported in many cases, thus the next step involves further improving efficacy and reducing toxicity as well as testing combination strategies with approved agents.
Collapse
Affiliation(s)
| | | | - Muhammad J. Niaz
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Aileen Lee
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Charlene Thomas
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Amy L. Hackett
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Priyanka Patel
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Naureen Rashid
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Scott T. Tagawa
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Department of Urology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
159
|
Zimmers SM, Browne EP, Williams KE, Jawale RM, Otis CN, Schneider SS, Arcaro KF. TROP2 methylation and expression in tamoxifen-resistant breast cancer. Cancer Cell Int 2018; 18:94. [PMID: 30002602 PMCID: PMC6034260 DOI: 10.1186/s12935-018-0589-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 06/21/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The DNA methyltransferase 1 inhibitor, 5-Aza-2'-deoxycytidine (5-Aza-dC) is a potential treatment for breast cancer. However, not all breast tumors will respond similarly to treatment with 5-Aza-dC, and little is known regarding the response of hormone-resistant breast cancers to 5-Aza-dC. METHODS We demonstrate that 5-Aza-dC-treatment has a stronger effect on an estrogen receptor-negative, Tamoxifen-selected cell line, TMX2-28, than on the estrogen receptor-positive, MCF7, parental cell line. Using data obtained from the HM450 Methylation Bead Chip, pyrosequencing, and RT-qPCR, we identified a panel of genes that are silenced by promoter methylation in TMX2-28 and re-expressed after treatment with 5-Aza-dC. RESULTS One of the genes identified, tumor associated calcium signal transducer 2 (TACSTD2), is altered by DNA methylation, and there is evidence that in some cancers decreased expression may result in greater proliferation. Analysis of DNA methylation of TACSTD2 and protein expression of its product, trophoblast antigen protein 2 (TROP2), was extended to a panel of primary (n = 34) and recurrent (n = 34) breast tumors. Stratifying tumors by both recurrence and ER status showed no significant relationship between TROP2 levels and TACSTD2 methylation. Knocking down TACSTD2 expression in MCF7 increased proliferation however; re-expressing TACSTD2 in TMX2-28 did not inhibit proliferation, indicating that TACSTD2 re-expression alone was insufficient to explain the decreased proliferation observed after treatment with 5-Aza-dC. CONCLUSIONS These results illustrate the complexity of the TROP2 signaling network. However, TROP2 may be a valid therapeutic target for some cancers. Further studies are needed to identify biomarkers that indicate how TROP2 signaling affects tumor growth and whether targeting TROP2 would be beneficial to the patient.
Collapse
Affiliation(s)
- Stephanie M. Zimmers
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Life Sciences Laboratories, Room 540D, 240 Thatcher Road, Amherst, MA 01003 USA
| | - Eva P. Browne
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Life Sciences Laboratories, Room 540D, 240 Thatcher Road, Amherst, MA 01003 USA
| | - Kristin E. Williams
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Life Sciences Laboratories, Room 540D, 240 Thatcher Road, Amherst, MA 01003 USA
| | - Rahul M. Jawale
- Pathology Department, Baystate Medical Center, 759 Chestnut Street, Springfield, MA 01199 USA
| | - Christopher N. Otis
- Pathology Department, Baystate Medical Center, 759 Chestnut Street, Springfield, MA 01199 USA
| | - Sallie S. Schneider
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Life Sciences Laboratories, Room 540D, 240 Thatcher Road, Amherst, MA 01003 USA
- Biospecimen Resource and Molecular Analysis Facility, Baystate Medical Center, 3601 Main Street, Springfield, MA 01199 USA
| | - Kathleen F. Arcaro
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, Life Sciences Laboratories, Room 540D, 240 Thatcher Road, Amherst, MA 01003 USA
| |
Collapse
|
160
|
Goldenberg DM, Stein R, Sharkey RM. The emergence of trophoblast cell-surface antigen 2 (TROP-2) as a novel cancer target. Oncotarget 2018; 9:28989-29006. [PMID: 29989029 PMCID: PMC6034748 DOI: 10.18632/oncotarget.25615] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/31/2018] [Indexed: 12/31/2022] Open
Abstract
TROP-2 is a glycoprotein first described as a surface marker of trophoblast cells, but subsequently shown to be increased in many solid cancers, with lower expression in certain normal tissues. It regulates cancer growth, invasion and spread by several signaling pathways, and has a role in stem cell biology and other diseases. This review summarizes TROP-2's properties, especially in cancer, and particularly its role as a target for antibody-drug conjugates (ADC) or immunotherapy. When the irinotecan metabolite, SN-38, is conjugated to a humanized anti-TROP-2 antibody (sacituzumab govitecan), it shows potent broad anticancer activity in human cancer xenografts and in patients with advanced triple-negative breast, non-small cell and small-cell lung, as well as urothelial cancers.
Collapse
Affiliation(s)
- David M. Goldenberg
- Center for Molecular Medicine and Immunology, Belleville, NJ, USA
- IBC Pharmaceuticals, Inc., Morris Plains, NJ, USA
| | - Rhona Stein
- Center for Molecular Medicine and Immunology, Belleville, NJ, USA
| | - Robert M. Sharkey
- Center for Molecular Medicine and Immunology, Belleville, NJ, USA
- Immunomedics, Inc., Morris Plains, NJ, USA
| |
Collapse
|
161
|
Zhang L, Yang G, Zhang R, Dong L, Chen H, Bo J, Xue W, Huang Y. Curcumin inhibits cell proliferation and motility via suppression of TROP2 in bladder cancer cells. Int J Oncol 2018; 53:515-526. [PMID: 29901071 PMCID: PMC6017220 DOI: 10.3892/ijo.2018.4423] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/20/2018] [Indexed: 12/22/2022] Open
Abstract
Bladder cancer (BC) has become a serious health prob-lem and represents the second most commonly diagnosed urological tumor. Curcumin is a principal active natural component of turmeric and has long been used in Asia as a traditional herbal medicine. Curcumin suppresses cell growth in various types of cancer, including BC, by regulating numerous molecular signaling pathways. The human trophoblast cell surface antigen 2 (Trop2) belongs to the tumor-associated calcium signal transducer gene family. Trop2 has been described as a cancer driver and is deregulated in various types of cancer. However, whether Trop2 is involved in curcumin-induced BC cell inhibition remains to be elucidated. The present study hypothesized that Trop2 may be a promising target of curcumin in BC cells. It was found that Trop2 was closely involved in curcumin-induced cell proliferation suppression, mobility inhibition, apoptosis, and cell cycle arrest in BC cells. Curcumin decreased the expression of Trop2 and its downstream target cyclin E1, and increased the level of p27. The overexpression of Trop2 enhanced the oncogenic activity of BC cells, whereas downregulation of the expression of Trop2 suppressed cell proliferation and mobility, increased apoptosis, and sensitized BC cells to curcumin treatment. Therefore, Trop2 may be a promising target of curcumin in BC cells and the inhibition of Trop2 may be an important method for the therapeutic management of patients with BC.
Collapse
Affiliation(s)
- Lianhua Zhang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Guoliang Yang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Ruiyun Zhang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Liang Dong
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Haige Chen
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Juanjie Bo
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Wei Xue
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Yiran Huang
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| |
Collapse
|
162
|
King GT, Eaton KD, Beagle BR, Zopf CJ, Wong GY, Krupka HI, Hua SY, Messersmith WA, El-Khoueiry AB. A phase 1, dose-escalation study of PF-06664178, an anti-Trop-2/Aur0101 antibody-drug conjugate in patients with advanced or metastatic solid tumors. Invest New Drugs 2018; 36:836-847. [PMID: 29333575 DOI: 10.1007/s10637-018-0560-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 01/02/2018] [Indexed: 12/16/2022]
Abstract
Purpose and Methods Trop-2 is a glycoprotein over-expressed in many solid tumors but at low levels in normal human tissue, providing a potential therapeutic target. We conducted a phase 1 dose-finding study of PF-06664178, an antibody-drug conjugate that targets Trop-2 for the selective delivery of the cytotoxic payload Aur0101. The primary objective was to determine the maximum tolerated dose and recommended phase 2 dose. Secondary objectives included further characterization of the safety profile, pharmacokinetics and antitumor activity. Eligible patients were enrolled and received multiple escalating doses of PF-06664178 in an open-label and unblinded manner based on a modified continual reassessment method. Results Thirty-one patients with advanced or metastatic solid tumors were treated with escalating doses of PF-06664178 given intravenously every 21 days. Doses explored ranged from 0.15 mg/kg to 4.8 mg/kg. Seven patients experienced at least one dose limiting toxicity (DLT), either neutropenia or rash. Doses of 3.60 mg/kg, 4.2 mg/kg and 4.8 mg/kg were considered intolerable due to DLTs in skin rash, mucosa and neutropenia. Best overall response was stable disease in 11 patients (37.9%). None of the patients had a partial or complete response. Systemic exposure of PF-06664178 increased in a dose-related manner. Serum concentrations of free Aur0101 were substantially lower than those of PF-06664178 and total antibody. No correlation of Trop-2 expression and objective response was observed, although Trop-2 overexpression was not required for study entry. The intermediate dose of 2.4 mg/kg appeared to be the highest tolerated dose, but this was not fully explored as the study was terminated early due to excess toxicity. Conclusion PF-06664178 showed toxicity at high dose levels with modest antitumor activity. Neutropenia, skin rash and mucosal inflammation were dose limiting toxicities. Findings from this study may potentially aid in future antibody drug conjugate design and trials.
Collapse
Affiliation(s)
- Gentry T King
- University of Colorado Cancer Center, Aurora, CO, USA.
| | - Keith D Eaton
- University of Washington Seattle Cancer Care Alliance, Seattle, WA, USA
| | - Brandon R Beagle
- Pfizer Early Oncology Development and Clinical Research, La Jolla, CA, USA
| | - Christopher J Zopf
- Pfizer Early Oncology Development and Clinical Research, La Jolla, CA, USA
| | - Gilbert Y Wong
- Pfizer Early Oncology Development and Clinical Research, South San Francisco, CA, USA
| | - Heike I Krupka
- Pfizer Early Oncology Development and Clinical Research, South San Francisco, CA, USA
| | - Steven Y Hua
- Pfizer Early Oncology Development and Clinical Research, La Jolla, CA, USA
| | | | - Anthony B El-Khoueiry
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| |
Collapse
|
163
|
Xu P, Zhao Y, Liu K, Lin S, Liu X, Wang M, Yang P, Tian T, Zhu YY, Dai Z. Prognostic role and clinical significance of trophoblast cell surface antigen 2 in various carcinomas. Cancer Manag Res 2017; 9:821-837. [PMID: 29276405 PMCID: PMC5731441 DOI: 10.2147/cmar.s147033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Trophoblast cell surface antigen 2 (TROP2) has been linked to disease prognosis in various human cancers and plays a critical role in tumor development, progression, and metastasis. A number of relevant studies have been published on this topic. A meta-analysis of the latest literature to evaluate the value of TROP2 as a predictive prognosticator of cancer was performed. METHODS Several online databases were searched, and relevant articles were retrieved. Overall and subcategory meta-analyses were performed, and results were collated. RESULTS Twenty-seven articles, including 29 studies, were included, involving 4,852 cancer patients, and results showed that the above-baseline expression of TROP2 was significantly associated with poorer overall survival (OS) (pooled hazard ratio [HR]: 1.84, 95% confidence interval [CI]: 1.45-2.35), disease-free survival (DFS) (pooled HR: 2.77, 95% CI: 1.73-4.42), and progression-free survival (PFS) (pooled HR: 1.71, 95% CI: 1.25-2.35). The following clinical characteristics were also significantly linked with TROP2 overexpression: moderate/poor differentiation (pooled HR: 3.03, 95% CI: 1.99-4.63), distant metastasis (pooled HR: 2.46, 95% CI: 1.05-5.75), lymph node metastasis (pooled HR: 2.47, 95%: CI 1.72-3.56), and advanced TNM stage (pooled HR: 2.02, 95% CI: 1.38-2.95). CONCLUSION TROP2 overexpression was predictive of poor prognosis in human cancers and may be an independent prognostic predictive biomarker. Further studies should be performed to confirm the significance of TROP2 in clinical practice.
Collapse
Affiliation(s)
- Peng Xu
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yang Zhao
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Kang Liu
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Shuai Lin
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Xinghan Liu
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Meng Wang
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Pengtao Yang
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Tian Tian
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yu-yao Zhu
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Zhijun Dai
- Department of Oncology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
164
|
Anampa J, Sparano JA. New agents for the management of resistant metastatic breast cancer. Expert Opin Pharmacother 2017; 18:1815-1831. [DOI: 10.1080/14656566.2017.1409206] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Jesus Anampa
- Department of Oncology, Section of Breast Medical Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Joseph A. Sparano
- Department of Oncology, Section of Breast Medical Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
165
|
Antibody–Drug Conjugates for the Treatment of Solid Tumors: Clinical Experience and Latest Developments. Target Oncol 2017; 12:719-739. [DOI: 10.1007/s11523-017-0535-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
166
|
Hochnadel I, Kossatz-Boehlert U, Jedicke N, Lenzen H, Manns MP, Yevsa T. Cancer vaccines and immunotherapeutic approaches in hepatobiliary and pancreatic cancers. Hum Vaccin Immunother 2017; 13:2931-2952. [PMID: 29112462 PMCID: PMC5718787 DOI: 10.1080/21645515.2017.1359362] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatobiliary and pancreatic cancers along with other gastrointestinal malignancies remain the leading cause of cancer-related deaths worldwide. Strategies developed in the recent years on immunotherapy and cancer vaccines in the setting of primary liver cancer as well as in pancreatic cancer are the scope of this review. Significance of orthotopic and autochthonous animal models which mimic and/or closely reflect human malignancies allowing for a prompt and trustworthy analysis of new therapeutics is underlined. Combinational approaches that on one hand, specifically target a defined cancer-driving pathway, and on the other hand, restore the functions of immune cells, which effector functions are often suppressed by a tumor milieu, are shown to have the strongest perspectives and future directions. Among combinational immunotherapeutic approaches a personalized- and individual cancer case-based therapy is of special importance.
Collapse
Affiliation(s)
- Inga Hochnadel
- a Department of Gastroenterology , Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany
| | - Uta Kossatz-Boehlert
- b Institute for Neuroanatomy, Eberhard-Karls University Tuebingen , Tuebingen , Germany
| | - Nils Jedicke
- a Department of Gastroenterology , Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany
| | - Henrike Lenzen
- a Department of Gastroenterology , Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany
| | - Michael P Manns
- a Department of Gastroenterology , Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany
| | - Tetyana Yevsa
- a Department of Gastroenterology , Hepatology and Endocrinology, Hannover Medical School , Hannover , Germany
| |
Collapse
|
167
|
Recent Advances in ADAM17 Research: A Promising Target for Cancer and Inflammation. Mediators Inflamm 2017; 2017:9673537. [PMID: 29230082 PMCID: PMC5688260 DOI: 10.1155/2017/9673537] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/15/2017] [Accepted: 09/11/2017] [Indexed: 02/06/2023] Open
Abstract
Since its discovery, ADAM17, also known as TNFα converting enzyme or TACE, is now known to process over 80 different substrates. Many of these substrates are mediators of cancer and inflammation. The field of ADAM metalloproteinases is at a crossroad with many of the new potential therapeutic agents for ADAM17 advancing into the clinic. Researchers have now developed potential drugs for ADAM17 that are selective and do not have the side effects which were seen in earlier chemical entities that targeted this enzyme. ADAM17 inhibitors have broad therapeutic potential, with properties ranging from tumor immunosurveillance and overcoming drug and radiation resistance in cancer, as treatments for cardiac hypertrophy and inflammatory conditions such as inflammatory bowel disease and rheumatoid arthritis. This review focuses on substrates and inhibitors identified more recently for ADAM17 and their role in cancer and inflammation.
Collapse
|
168
|
Trop2 enhances invasion of thyroid cancer by inducing MMP2 through ERK and JNK pathways. BMC Cancer 2017; 17:486. [PMID: 28709407 PMCID: PMC5513028 DOI: 10.1186/s12885-017-3475-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 07/06/2017] [Indexed: 12/18/2022] Open
Abstract
Background Mounting evidence has showed that Tumor-associated calcium signal transducer 2 (Trop2) is upregulated in various kinds of human cancers and plays important roles in tumorigenesis. However, the expression status and functional significance of Trop2 in thyroid cancer are largely unknown. Methods We first determined the expression of Trop2 by using RNAseqV2 data sets for thyroid cancer deposited on The Cancer Genome Atlas (TCGA) website. The expression of Trop2 was then confirmed by real-time reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry assays. Cell invasion and migration were assessed by conducting Transwell and wound healing assays. Furthermore, we explored the underlying mechanisms by using real-time RT-PCR, Western blot, zymography, and luciferase reporter assays. Results In this study, we demonstrated that the expression of Trop2 was significantly elevated in thyroid cancer and that its expression level was correlated with the tumor-node-metastasis (TNM) staging and N classification. Dysregulation of Trop2 altered the invasive capability of thyroid cancer cells. Further mechanistic study revealed that MMP2 expression was upregulated by Trop2. Moreover, we found that the effects of Trop2 were dependent on ERK and JNK pathways. The results from clinical specimens showed that Trop2 expression correlated with MMP2 expression in primary thyroid cancer. Conclusion The current study suggests that elevated expression of Trop2 may represent an important molecular hallmark that is biologically and clinically relevant to the progression of thyroid cancer.
Collapse
|
169
|
Bardia A, Mayer IA, Diamond JR, Moroose RL, Isakoff SJ, Starodub AN, Shah NC, O’Shaughnessy J, Kalinsky K, Guarino M, Abramson V, Juric D, Tolaney SM, Berlin J, Messersmith WA, Ocean AJ, Wegener WA, Maliakal P, Sharkey RM, Govindan SV, Goldenberg DM, Vahdat LT. Efficacy and Safety of Anti-Trop-2 Antibody Drug Conjugate Sacituzumab Govitecan (IMMU-132) in Heavily Pretreated Patients With Metastatic Triple-Negative Breast Cancer. J Clin Oncol 2017. [DOI: 10.1200/jco.2016.70.8297.2017.2.test] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Aditya Bardia
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Ingrid A. Mayer
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Jennifer R. Diamond
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Rebecca L. Moroose
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Steven J. Isakoff
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Alexander N. Starodub
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Nikita C. Shah
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Joyce O’Shaughnessy
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Kevin Kalinsky
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Michael Guarino
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Vandana Abramson
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Dejan Juric
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Sara M. Tolaney
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Jordan Berlin
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Wells A. Messersmith
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Allyson J. Ocean
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - William A. Wegener
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Pius Maliakal
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Robert M. Sharkey
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Serengulam V. Govindan
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - David M. Goldenberg
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| | - Linda T. Vahdat
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida
| |
Collapse
|
170
|
Ocean AJ, Starodub AN, Bardia A, Vahdat LT, Isakoff SJ, Guarino M, Messersmith WA, Picozzi VJ, Mayer IA, Wegener WA, Maliakal P, Govindan SV, Sharkey RM, Goldenberg DM. Sacituzumab govitecan (IMMU-132), an anti-Trop-2-SN-38 antibody-drug conjugate for the treatment of diverse epithelial cancers: Safety and pharmacokinetics. Cancer 2017; 123:3843-3854. [DOI: 10.1002/cncr.30789] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/29/2017] [Accepted: 04/19/2017] [Indexed: 12/27/2022]
Affiliation(s)
| | | | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Harvard Medical School; Boston Massachusetts
| | | | - Steven J. Isakoff
- Massachusetts General Hospital Cancer Center, Harvard Medical School; Boston Massachusetts
| | | | | | | | | | | | | | | | | | | |
Collapse
|
171
|
Heist RS, Guarino MJ, Masters G, Purcell WT, Starodub AN, Horn L, Scheff RJ, Bardia A, Messersmith WA, Berlin J, Ocean AJ, Govindan SV, Maliakal P, Mudenda B, Wegener WA, Sharkey RM, Goldenberg DM, Camidge DR. Therapy of Advanced Non-Small-Cell Lung Cancer With an SN-38-Anti-Trop-2 Drug Conjugate, Sacituzumab Govitecan. J Clin Oncol 2017; 35:2790-2797. [PMID: 28548889 DOI: 10.1200/jco.2016.72.1894] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Purpose Trop-2, expressed in most solid cancers, may be a target for antibody-drug conjugates (ADCs) in non-small-cell lung cancer (NSCLC). We studied sacituzumab govitecan (IMMU-132), a Trop-2 ADC, for the targeting of SN-38. Patients and Methods We evaluated IMMU-132 in a single-arm multicenter trial in patients with pretreated metastatic NSCLC who received either 8 or 10 mg/kg on days 1 and 8 of 21-day cycles. The primary end points were safety and objective response rate (ORR). Progression-free survival and overall survival were secondary end points. Results Fifty-four patients were treated. In the response-assessable study population (n = 47), which had a median of three prior therapies (range, two to seven), the ORR was 19%; median response duration, 6.0 months (95% CI, 4.8 to 8.3 months); and clinical benefit rate (complete response + partial response + stable disease ≥ 4 months), 43%. ORR in the intention-to-treat (ITT) population was 17% (nine of 54). Responses occurred with a median onset of 3.8 months, including patients who had relapsed or progressed after immune checkpoint inhibitor therapy. Median ITT progression-free survival was 5.2 months (95% CI, 3.2 to 7.1 months) and median ITT overall survival, 9.5 months (95% CI, 5.9 to 16.7 months). Grade 3 or higher adverse events included neutropenia (28%), diarrhea (7%), nausea (7%), fatigue (6%), and febrile neutropenia (4%). One patient developed a transient immune response, despite patients receiving a median of 10 doses. More than 90% of 26 assessable archival tumor specimens were highly positive (2+, 3+) for Trop-2 by immunohistochemistry, which suggests that Trop-2 is not a predictive biomarker for response. Conclusion IMMU-132 was well-tolerated and induced durable responses in heavily pretreated patients with metastatic NSCLC. This ADC should be studied further in this disease and in other patients with Trop-2-expressing tumors.
Collapse
Affiliation(s)
- Rebecca Suk Heist
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Michael J Guarino
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Gregory Masters
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - W Thomas Purcell
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Alexander N Starodub
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Leora Horn
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Ronald J Scheff
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Aditya Bardia
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Wells A Messersmith
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Jordan Berlin
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Allyson J Ocean
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Serengulam V Govindan
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Pius Maliakal
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Boyd Mudenda
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - William A Wegener
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Robert M Sharkey
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - David M Goldenberg
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - D Ross Camidge
- Rebecca Suk Heist and Aditya Bardia, Massachusetts General Hospital Cancer Center; Rebecca Suk Heist and Aditya Bardia, Harvard Medical School, Boston, MA; Michael J. Guarino and Gregory Masters, Helen F. Graham Cancer Center & Research Institute, Newark, DE; W. Thomas Purcell, Wells A. Messersmith, and D. Ross Camidge, University of Colorado Cancer Center, Aurora, CO; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Leora Horn and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Ronald J. Scheff and Allyson J. Ocean, Weill Cornell Medicine, New York, NY; and Serengulam V. Govindan, Pius Maliakal, Boyd Mudenda, William A. Wegener, Robert M. Sharkey, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| |
Collapse
|
172
|
Abstract
INTRODUCTION Despite advances in the diagnosis and treatment of patients with cancer, patients with metastatic cancer have limited therapeutic options after initial lines of therapy. Understanding tumor biology has translated into the identification of actionable targets that resulted in therapeutics. Antibody-drug conjugates (ADC) are capitalizing on this explosion of scientific information. ADCs allow an antibody to a unique target to be conjugated via an innovative linker, to a highly toxic drug which is delivered to its target. Sacituzumab govitecan is an ADC that combines the active molecule in irinotecan, SN-38, to an antibody targeting trop2. Areas covered: In this review, the authors introduce the reader to the ADC sacituzumab govitecan providing the reader with details about its pharmacokinetics, pharmacodynamics, efficacy and safety. The authors also give their expert analysis about its potential future use. Expert opinion: Sacituzumab govitecan is a novel and well-tolerated therapeutic showing promising results in difficult to treat cancers. Further studies are underway to optimize the group of patients that would benefit from it. Given its excellent performance, we are cautiously optimistic it will be approved by the FDA.
Collapse
Affiliation(s)
- Sheena Sahota
- a Department of Medicine , Weill Cornell Medicine , New York , NY , USA
| | - Linda T Vahdat
- a Department of Medicine , Weill Cornell Medicine , New York , NY , USA
| |
Collapse
|
173
|
Sav A, Rotondo F, Syro LV, Altinoz MA, Kovacs K. Selective molecular biomarkers to predict biologic behavior in pituitary tumors. Expert Rev Endocrinol Metab 2017; 12:177-185. [PMID: 30063456 DOI: 10.1080/17446651.2017.1312341] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
To date, several cell proliferation markers, apoptosis, vascular markers, oncogenes, tumor suppressor genes, cell cycle mediators, microRNA (miRNAs), and long noncoding RNAs (lncRNAs) have been identified to be involved in the tumorigenesis, migration, proliferation and invasiveness of pituitary adenomas. There are still no reliable morphologic markers predictive of pituitary adenoma recurrence. Recent scientific research introduced new techniques to enable us to attain new information on the genesis and biologic behavior of pituitary adenomas. Areas covered: This review covers selected, compelling and cumulative information in regards to TACSTD family (EpCAM, TROP2), neuropilin (NRP-1), oncogene-induced senescence (OIS), fascins (FSCN1), invasion-associated genes (CLDN7, CNTNAP2, ITGA6, JAM3, PTPRC and CTNNA1) EZH2, and ENC1 genes and endocan. Expert commentary: Ongoing research provides clinicians, surgeons and researchers with new information not only on diverse pathways in tumorigenesis but also on the clinical aggressive behavior of pituitary adenomas. Newly developed molecular techniques, bioinformatics and new pharmaceutical drug options are helpful tools to widen the perspectives in our understanding of the complex nature of pituitary tumorigenesis. The discovery of new molecular biomarkers can only be accomplished by continuing to investigate pituitary embryogenesis, histogenesis and tumorigenesis.
Collapse
Affiliation(s)
- Aydin Sav
- a Division of Neuropathology , Nisantasi Pathology Group , Istanbul , Turkey
| | - Fabio Rotondo
- b Department of Laboratory Medicine, Division of Pathology, St. Michael's Hospital , University of Toronto , Toronto , ON , Canada
| | - Luis V Syro
- c Department of Neurosurgery , Hospital Pablo Tobon Uribe and Clinica Medellin , Medellin , Colombia
| | - Meric A Altinoz
- d Department of Immunology, Experimental Medical Research Institute , Istanbul University , Istanbul , Turkey
| | - Kalman Kovacs
- b Department of Laboratory Medicine, Division of Pathology, St. Michael's Hospital , University of Toronto , Toronto , ON , Canada
| |
Collapse
|
174
|
Avellini C, Licini C, Lazzarini R, Gesuita R, Guerra E, Tossetta G, Castellucci C, Giannubilo SR, Procopio A, Alberti S, Mazzucchelli R, Olivieri F, Marzioni D. The trophoblast cell surface antigen 2 and miR-125b axis in urothelial bladder cancer. Oncotarget 2017; 8:58642-58653. [PMID: 28938585 PMCID: PMC5601681 DOI: 10.18632/oncotarget.17407] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 03/30/2017] [Indexed: 11/29/2022] Open
Abstract
Human trophoblast cell surface antigen 2 (Trop-2) is a 40-kDa transmembrane glycoprotein that was first identified as a marker of human trophoblast cells. Trop-2 acts on cell proliferation, adhesion, and migration by activating a number of intracellular signalling pathways. Elevated Trop-2 expression has been demonstrated in several types of cancer and correlated with aggressiveness and poor prognosis. Since no data are available on Trop-2 in bladder cancer (BC), the purpose of the study was to determine its levels in tissue specimens from normal individuals and patients with BC at different stages. Moreover, since according to recent evidence Trop-2 is a miR-125b target, miR-125b expression was also assessed in tissue specimens. Finally, the effect of the Trop-2/miR-125b axis on the proliferation and migration of BC cells was evaluated in vitro. The Trop-2/miR-125b axis was seen to be differentially expressed in normal urothelium, non-invasive BC and invasive BC tissue. Significant miR-125b down-regulation was associated with a significant increase in Trop-2 protein levels in BC tissue and correlated with disease severity. In vitro analysis confirmed the role of miR-125b in down-modulation of Trop-2 protein levels and showed that Trop-2/miR-125b axis affects cellular proliferation in bladder tissue. In conclusion, our findings highlight a role for the Trop-2/miR-125b axis in BC progression and suggest Trop-2 and miR-125b as diagnostic/prognostic marker candidates as well as druggable targets for innovative therapeutic approaches.
Collapse
Affiliation(s)
- Chiara Avellini
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Caterina Licini
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Raffaella Lazzarini
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Rosaria Gesuita
- Centre of Epidemiology, Biostatistics and Medical Information Technology, Università Politecnica delle Marche, Ancona, Italy
| | - Emanuela Guerra
- Unit of Cancer Pathology, CeSI-MeT, "G. d'Annunzio" University, Chieti, Italy.,ONCOXX Biotech SRL, Chieti, Italy
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Clara Castellucci
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | | | - Antonio Procopio
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy.,Center of Clinical Pathology and Innovative Therapy, National Institute INRCA-IRCCS, Ancona, Italy
| | - Saverio Alberti
- Unit of Cancer Pathology, CeSI-MeT, "G. d'Annunzio" University, Chieti, Italy.,ONCOXX Biotech SRL, Chieti, Italy
| | - Roberta Mazzucchelli
- Section of Pathological Anatomy, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, United Hospitals, Ancona, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy.,Center of Clinical Pathology and Innovative Therapy, National Institute INRCA-IRCCS, Ancona, Italy
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
175
|
Bardia A, Mayer IA, Diamond JR, Moroose RL, Isakoff SJ, Starodub AN, Shah NC, O'Shaughnessy J, Kalinsky K, Guarino M, Abramson V, Juric D, Tolaney SM, Berlin J, Messersmith WA, Ocean AJ, Wegener WA, Maliakal P, Sharkey RM, Govindan SV, Goldenberg DM, Vahdat LT. Efficacy and Safety of Anti-Trop-2 Antibody Drug Conjugate Sacituzumab Govitecan (IMMU-132) in Heavily Pretreated Patients With Metastatic Triple-Negative Breast Cancer. J Clin Oncol 2017; 35:2141-2148. [PMID: 28291390 DOI: 10.1200/jco.2016.70.8297] [Citation(s) in RCA: 285] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose Trop-2, expressed in most triple-negative breast cancers (TNBCs), may be a potential target for antibody-drug conjugates. Sacituzumab govitecan, an antibody-drug conjugate, targets Trop-2 for the selective delivery of SN-38, the active metabolite of irinotecan. Patients and Methods We evaluated sacituzumab govitecan in a single-arm, multicenter trial in patients with relapsed/refractory metastatic TNBC who received a 10 mg/kg starting dose on days 1 and 8 of 21-day repeated cycles. The primary end points were safety and objective response rate; secondary end points were progression-free survival and overall survival. Results In 69 patients who received a median of five prior therapies (range, one to 12) since diagnosis, the confirmed objective response rate was 30% (partial response, n = 19; complete response, n = 2), the median response duration was 8.9 (95% CI, 6.1 to 11.3) months, and the clinical benefit rate (complete response + partial response + stable disease ≥ 6 months) was 46%. These responses occurred early, with a median onset of 1.9 months. Median progression-free survival was 6.0 (95% CI, 5.0 to 7.3) months, and median overall survival was 16.6 (95% CI, 11.1 to 20.6) months. Grade ≥ 3 adverse events included neutropenia (39%), leukopenia (16%), anemia (14%), and diarrhea (13%); the incidence of febrile neutropenia was 7%. The majority of archival tumor specimens (88%) were moderately to strongly positive for Trop-2 by immunohistochemistry. No neutralizing antibodies to the ADC or antibody were detected, despite repeated cycles developed. Conclusion Sacituzumab govitecan was well tolerated and induced early and durable responses in heavily pretreated patients with metastatic TNBC. As a therapeutic target and predictive biomarker, Trop-2 warrants further research.
Collapse
Affiliation(s)
- Aditya Bardia
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Ingrid A Mayer
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Jennifer R Diamond
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Rebecca L Moroose
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Steven J Isakoff
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Alexander N Starodub
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Nikita C Shah
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Joyce O'Shaughnessy
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Kevin Kalinsky
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Michael Guarino
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Vandana Abramson
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Dejan Juric
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Sara M Tolaney
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Jordan Berlin
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Wells A Messersmith
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Allyson J Ocean
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - William A Wegener
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Pius Maliakal
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Robert M Sharkey
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Serengulam V Govindan
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - David M Goldenberg
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| | - Linda T Vahdat
- Aditya Bardia, Steven J. Isakoff, and Dejan Juric, Massachusetts General Hospital Cancer Center; Aditya Bardia, Steven J. Isakoff, Dejan Juric, and Sara M. Tolaney, Harvard Medical School; Sara M. Tolaney, Dana-Farber Cancer Institute, Boston, MA; Ingrid A. Mayer, Vandana Abramson, and Jordan Berlin, Vanderbilt-Ingram Cancer Center, Nashville, TN; Jennifer R. Diamond and Wells A. Messersmith, University of Colorado Cancer Center, Aurora, CO; Rebecca L. Moroose and Nikita C. Shah, University of Florida Health Cancer Center, Orlando, FL; Alexander N. Starodub, Indiana University Health Center for Cancer Care, Goshen, IN; Joyce O'Shaughnessy, Texas Oncology-Baylor Charles A. Sammons Cancer Center; Joyce O'Shaughnessy, US Oncology, Dallas, TX; Kevin Kalinsky, Columbia University Herbert Irving Comprehensive Cancer Center; Allyson J. Ocean and Linda T. Vahdat, Weill Cornell Medicine, New York, NY; Michael Guarino, Helen F. Graham Cancer Center, Newark, DE; William A. Wegener, Pius Maliakal, Robert M. Sharkey, Serengulam V. Govindan, and David M. Goldenberg, Immunomedics, Morris Plains, NJ
| |
Collapse
|
176
|
Cheng N, Li H, Luo J. Trop2 promotes proliferation, invasion and EMT of nasopharyngeal carcinoma cells through the NF-κB pathway. RSC Adv 2017. [DOI: 10.1039/c7ra09915k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Human trophoblast cell surface antigen 2 (Trop2), a cell surface transmembrane glycoprotein receptor, has been demonstrated to be closely associated with increasing tumor aggressiveness, metastasis and unfavorable prognosis.
Collapse
Affiliation(s)
- Nan Cheng
- Department of Otolaryngological
- Huaihe Hospital of Henan University
- Kaifeng
- China
| | - Haixia Li
- Department of Otolaryngological
- Huaihe Hospital of Henan University
- Kaifeng
- China
| | - Junpeng Luo
- Department of Oncology
- Huaihe Hospital of Henan University
- Kaifeng
- China
| |
Collapse
|
177
|
Zeng P, Chen MB, Zhou LN, Tang M, Liu CY, Lu PH. Impact of TROP2 expression on prognosis in solid tumors: A Systematic Review and Meta-analysis. Sci Rep 2016; 6:33658. [PMID: 27645103 PMCID: PMC5028884 DOI: 10.1038/srep33658] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/30/2016] [Indexed: 01/20/2023] Open
Abstract
Over-expression of TROP2 (the trophoblast cell surface antigen 2) was reported to predict poor prognosis in various solid tumors in number of studies. However, the results remained not comprehensive. Therefore, we here carried out this meta-analysis of relevant studies published on this topic to quantitatively evaluate the clinicopathological significance of TROP2 in solid tumors. Relevant articles were identified through searching the PubMed, Web of Science and Embase database. The primary outcomes were overall survival (OS) and disease-free survival (DFS). In this meta-analysis, 16 studies involving 2,569 participants were included, and we drew the conclusion that TROP2 overexpression was significantly associated with poor OS (pooled HR = 1.896, 95% CI = 1.599–2.247, P < 0.001) and short DFS (pooled HR = 2.336, 95% CI = 1.596–3.419, P < 0.001). Furthermore, the subgroup analysis revealed that the associations between TROP2 overexpression and the outcome endpoints (OS or DFS) were significant in in patients with female genital system neoplasms, as well in gastrointestine neoplasms. In addition, subgroup analysis found no difference HR across populations of different descent.Taken together, TROP2 overexpression was associated with poor survival in human solid tumors. TROP2 may be a valuable prognosis predictive biomarker and a potential therapeutic target in human solid tumors.
Collapse
Affiliation(s)
- Ping Zeng
- Department of Radiotherapy and Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, 91 Qianjin Road, Kunshan 215300, Jiangsu Province, China
| | - Min-Bin Chen
- Department of Radiotherapy and Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, 91 Qianjin Road, Kunshan 215300, Jiangsu Province, China
| | - Li-Na Zhou
- Department of Radiotherapy and Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, 91 Qianjin Road, Kunshan 215300, Jiangsu Province, China
| | - Min Tang
- Department of Radiotherapy and Oncology, Kunshan First People's Hospital Affiliated to Jiangsu University, 91 Qianjin Road, Kunshan 215300, Jiangsu Province, China
| | - Chao-Ying Liu
- Department of Medical Oncology, Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi 214023, Jiangsu Province, China
| | - Pei-Hua Lu
- Department of Medical Oncology, Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi 214023, Jiangsu Province, China
| |
Collapse
|
178
|
Systems-level effects of ectopic galectin-7 reconstitution in cervical cancer and its microenvironment. BMC Cancer 2016; 16:680. [PMID: 27558259 PMCID: PMC4997669 DOI: 10.1186/s12885-016-2700-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 08/09/2016] [Indexed: 12/20/2022] Open
Abstract
Background Galectin-7 (Gal-7) is negatively regulated in cervical cancer, and appears to be a link between the apoptotic response triggered by cancer and the anti-tumoral activity of the immune system. Our understanding of how cervical cancer cells and their molecular networks adapt in response to the expression of Gal-7 remains limited. Methods Meta-analysis of Gal-7 expression was conducted in three cervical cancer cohort studies and TCGA. In silico prediction and bisulfite sequencing were performed to inquire epigenetic alterations. To study the effect of Gal-7 on cervical cancer, we ectopically re-expressed it in the HeLa and SiHa cervical cancer cell lines, and analyzed their transcriptome and SILAC-based proteome. We also examined the tumor and microenvironment host cell transcriptomes after xenotransplantation into immunocompromised mice. Differences between samples were assessed with the Kruskall-Wallis, Dunn’s Multiple Comparison and T tests. Kaplan–Meier and log-rank tests were used to determine overall survival. Results Gal-7 was constantly downregulated in our meta-analysis (p < 0.0001). Tumors with combined high Gal-7 and low galectin-1 expression (p = 0.0001) presented significantly better prognoses (p = 0.005). In silico and bisulfite sequencing assays showed de novo methylation in the Gal-7 promoter and first intron. Cells re-expressing Gal-7 showed a high apoptosis ratio (p < 0.05) and their xenografts displayed strong growth retardation (p < 0.001). Multiple gene modules and transcriptional regulators were modulated in response to Gal-7 reconstitution, both in cervical cancer cells and their microenvironments (FDR < 0.05 %). Most of these genes and modules were associated with tissue morphogenesis, metabolism, transport, chemokine activity, and immune response. These functional modules could exert the same effects in vitro and in vivo, even despite different compositions between HeLa and SiHa samples. Conclusions Gal-7 re-expression affects the regulation of molecular networks in cervical cancer that are involved in diverse cancer hallmarks, such as metabolism, growth control, invasion and evasion of apoptosis. The effect of Gal-7 extends to the microenvironment, where networks involved in its configuration and in immune surveillance are particularly affected. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2700-8) contains supplementary material, which is available to authorized users.
Collapse
|
179
|
Zheng Z, Dong XJ. Clinical value of serum trophoblast cell surface protein 2 (TROP2) antibody in non-small-cell lung cancer patients. Biomarkers 2016; 21:739-742. [PMID: 27310709 DOI: 10.1080/1354750x.2016.1201532] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE This study was to explore the clinical role of serum trophoblast cell surface protein 2 (TROP2) antibody in patients with non-small-cell lung cancer (NSCLC). MATERIALS AND METHODS We collected serum specimens from 117 NSCLC patients, 40 benign lung disease patients, and 60 healthy controls. TROP2 antibody concentrations were measured using enzyme-linked immunosorbent assay. RESULTS Serum TROP2 antibody levels were higher in the NSCLC group compared to the control group (p < 0.001). TROP2 antibody, at a cutoff value of 9.8 U/ml, showed good diagnostic performance for NSCLC. CONCLUSION Measurement of TROP2 antibody might have diagnostic value for patients with NSCLC.
Collapse
Affiliation(s)
- Zhuan Zheng
- a Clinical Laboratory Center of Shaoxing People's Hospital , Shaoxing Hospital of Zhejiang University , Shaoxing , China
| | - Xue-Jun Dong
- a Clinical Laboratory Center of Shaoxing People's Hospital , Shaoxing Hospital of Zhejiang University , Shaoxing , China
| |
Collapse
|
180
|
Guerra E, Trerotola M, Tripaldi R, Aloisi AL, Simeone P, Sacchetti A, Relli V, D'Amore A, La Sorda R, Lattanzio R, Piantelli M, Alberti S. Trop-2 Induces Tumor Growth Through AKT and Determines Sensitivity to AKT Inhibitors. Clin Cancer Res 2016; 22:4197-205. [PMID: 27022065 DOI: 10.1158/1078-0432.ccr-15-1701] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 02/29/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Inhibition of AKT is a key target area for personalized cancer medicine. However, predictive markers of response to AKT inhibitors are lacking. Correspondingly, the AKT-dependent chain of command for tumor growth, which will mediate AKT-dependent therapeutic responses, remains unclear. EXPERIMENTAL DESIGN Proteomic profiling was utilized to identify nodal hubs of the Trop-2 cancer growth-driving network. Kinase-specific inhibitors were used to dissect Trop-2-dependent from Trop-2-independent pathways. In vitro assays, in vivo preclinical models, and case series of primary human breast cancers were utilized to define the mechanisms of Trop-2-driven growth and the mode of action of Trop-2-predicted AKT inhibitors. RESULTS Trop-2 and AKT expression was shown to be tightly coordinated in human breast cancers, with virtual overlap with AKT activation profiles at T308 and S473, consistent with functional interaction in vivo AKT allosteric inhibitors were shown to only block the growth of Trop-2-expressing tumor cells, both in vitro and in preclinical models, being ineffective on Trop-2-null cells. Consistently, AKT-targeted siRNA only impacted on Trop-2-expressing cells. Lentiviral downregulation of endogenous Trop-2 abolished tumor response to AKT blockade, indicating Trop-2 as a mandatory activator of AKT. CONCLUSIONS Our findings indicate that the expression of Trop-2 is a stringent predictor of tumor response to AKT inhibitors. They also support the identification of target-activatory pathways, as efficient predictors of response in precision cancer therapy. Clin Cancer Res; 22(16); 4197-205. ©2016 AACR.
Collapse
Affiliation(s)
- Emanuela Guerra
- Unit of Cancer Pathology, CeSI-MeT, University 'G. d'Annunzio,' Chieti, Italy
| | - Marco Trerotola
- Unit of Cancer Pathology, CeSI-MeT, University 'G. d'Annunzio,' Chieti, Italy
| | - Romina Tripaldi
- Unit of Cancer Pathology, CeSI-MeT, University 'G. d'Annunzio,' Chieti, Italy
| | - Anna Laura Aloisi
- Unit of Cancer Pathology, CeSI-MeT, University 'G. d'Annunzio,' Chieti, Italy
| | - Pasquale Simeone
- Unit of Cancer Pathology, CeSI-MeT, University 'G. d'Annunzio,' Chieti, Italy
| | - Andrea Sacchetti
- Unit of Cancer Pathology, CeSI-MeT, University 'G. d'Annunzio,' Chieti, Italy
| | - Valeria Relli
- Unit of Cancer Pathology, CeSI-MeT, University 'G. d'Annunzio,' Chieti, Italy
| | - Antonella D'Amore
- Unit of Cancer Pathology, CeSI-MeT, University 'G. d'Annunzio,' Chieti, Italy
| | - Rossana La Sorda
- Unit of Cancer Pathology, CeSI-MeT, University 'G. d'Annunzio,' Chieti, Italy
| | - Rossano Lattanzio
- Unit of Cancer Pathology, CeSI-MeT, University 'G. d'Annunzio,' Chieti, Italy. Department of Medical, Oral and Biotechnological Sciences, University 'G. d'Annunzio,' Chieti, Italy
| | - Mauro Piantelli
- Unit of Cancer Pathology, CeSI-MeT, University 'G. d'Annunzio,' Chieti, Italy. Department of Medical, Oral and Biotechnological Sciences, University 'G. d'Annunzio,' Chieti, Italy
| | - Saverio Alberti
- Unit of Cancer Pathology, CeSI-MeT, University 'G. d'Annunzio,' Chieti, Italy. Department of Neuroscience, Imaging and Clinical Sciences, Unit of Physiology and Physiopathology, University 'G. d'Annunzio,' Chieti, Italy.
| |
Collapse
|
181
|
Simms A, Jacob RP, Cohen C, Siddiqui MT. TROP-2 expression in papillary thyroid carcinoma. Diagn Cytopathol 2015; 44:26-31. [DOI: 10.1002/dc.23382] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/21/2015] [Accepted: 09/24/2015] [Indexed: 01/21/2023]
Affiliation(s)
- Anthony Simms
- Department of Pathology and Laboratory Medicine; Emory University Hospital; Atlanta Georgia
| | - Reuben P. Jacob
- Department of Pathology and Laboratory Medicine; Emory University Hospital; Atlanta Georgia
| | - Cynthia Cohen
- Department of Pathology and Laboratory Medicine; Emory University Hospital; Atlanta Georgia
| | - Momin T. Siddiqui
- Department of Pathology and Laboratory Medicine; Emory University Hospital; Atlanta Georgia
| |
Collapse
|
182
|
Gui T, Dong X, Li R, Li Y, Wang Z. Identification of hepatocellular carcinoma-related genes with a machine learning and network analysis. J Comput Biol 2015; 22:63-71. [PMID: 25247452 DOI: 10.1089/cmb.2014.0122] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Liver cancer is one of the leading causes of cancer mortality worldwide. Hepatocellular carcinoma (HCC) is the main type of liver cancer. We applied a machine learning approach with maximum-relevance-minimum-redundancy (mRMR) algorithm followed by incremental feature selection (IFS) to a set of microarray data generated from 43 tumor and 52 nontumor samples. With the machine learning approach, we identified 117 gene probes that could optimally separate tumor and nontumor samples. These genes not only include known HCC-relevant genes such as MT1X, BMI1, and CAP2, but also include cancer genes that were not found previously to be closely related to HCC, such as TACSTD2. Then, we constructed a molecular interaction network based on the protein-protein interaction (PPI) data from the STRING database and identified 187 genes on the shortest paths among the genes identified with the machine learning approach. Network analysis reveals new potential roles of ubiquitin C in the pathogenesis of HCC. Based on gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, we showed that the identified subnetwork is significantly enriched in biological processes related to cell death. These results bring new insights of understanding the process of HCC.
Collapse
Affiliation(s)
- Tuantuan Gui
- 1 Key Laboratory of Systems Biology, Shanghai Institutes for Biological Sciences , Chinese Academy of Sciences, Shanghai, P.R. China
| | | | | | | | | |
Collapse
|
183
|
Zhao P, Yu HZ, Cai JH. Clinical investigation of TROP-2 as an independent biomarker and potential therapeutic target in colon cancer. Mol Med Rep 2015; 12:4364-4369. [PMID: 26059528 DOI: 10.3892/mmr.2015.3900] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Accepted: 05/06/2015] [Indexed: 11/10/2022] Open
Abstract
Colon cancer is associated with a severe demographic and economic burden worldwide. The pathogenesis of colon cancer is highly complex and involves sequential genetic and epigenetic mechanisms. Despite extensive investigation, the pathogenesis of colon cancer remains to be elucidated. As the third most common type of cancer worldwide, the treatment options for colon cancer are currently limited. Human trophoblast cell‑surface marker (TROP‑2), is a cell‑surface transmembrane glycoprotein overexpressed by several types of epithelial carcinoma. In addition, TROP‑2 has been demonstrated to be associated with tumorigenesis and invasiveness in solid types of tumor. The aim of the present study was to investigate the protein expression of TROP‑2 in colon cancer tissues, and further explore the association between the expression of TROP‑2 and clinicopathological features of patients with colon cancer. The expression and localization of the TROP‑2 protein was examined using western blot analysis and immunofluorescence staining. Finally, the expression of TROP‑2 expression was correlated to conventional clinicopathological features of colon cancer using a χ2 test. The results revealed that TROP‑2 protein was expressed at high levels in the colon cancer tissues, which was associated with the development and pathological process of colon cancer. Therefore, TROP‑2 may be used as a biomarker to determine the clinical prognosis, and as a potential therapeutic target in colon cancer.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Hai-Zheng Yu
- Department of Social Sciences, Hebei United University, Tangshan, Hebei 063000, P.R. China
| | - Jian-Hui Cai
- Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| |
Collapse
|
184
|
Molecular targets and pathways involved in liver metastasis of colorectal cancer. Clin Exp Metastasis 2015; 32:623-35. [DOI: 10.1007/s10585-015-9732-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 06/17/2015] [Indexed: 02/08/2023]
|
185
|
Trop-2 is a novel target for solid cancer therapy with sacituzumab govitecan (IMMU-132), an antibody-drug conjugate (ADC). Oncotarget 2015; 6:22496-512. [PMID: 26101915 PMCID: PMC4673178 DOI: 10.18632/oncotarget.4318] [Citation(s) in RCA: 339] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 06/05/2015] [Indexed: 12/27/2022] Open
Abstract
Trop-2 is a novel target for ADC therapy because of its high expression by many solid cancers. The rational development of IMMU-132 represents a paradigm shift as an ADC that binds a well-known moderately-cytotoxic drug, SN-38, to the anti-Trop-2 antibody. In vitro and in vivo studies show enhanced efficacy, while there is a gradual release of SN-38 that contributes to the overall effect. IMMU-132 is most efficacious at a high drug:antibody ratio (DAR) of 7.6:1, which does not affect binding and pharmacokinetics. It targets up to 136-fold more SN-38 to a human cancer xenograft than irinotecan, SN-38's prodrug. IMMU-132 delivers SN-38 in its most active, non-glucuronidated form, which may explain the lower frequency of severe diarrhea than with irinotecan. Thus, this ADC, carrying a moderately-toxic drug targeting Trop-2 represents a novel cancer therapeutic that is showing promising activity in patients with several metastatic cancer types, including triple-negative breast cancer, non-small-cell and small-cell lung cancers.
Collapse
|
186
|
Lokody IB, Francis JC, Gardiner JR, Erler JT, Swain A. Pten Regulates Epithelial Cytodifferentiation during Prostate Development. PLoS One 2015; 10:e0129470. [PMID: 26076167 PMCID: PMC4468205 DOI: 10.1371/journal.pone.0129470] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 05/10/2015] [Indexed: 01/08/2023] Open
Abstract
Gene expression and functional studies have indicated that the molecular programmes involved in prostate development are also active in prostate cancer. PTEN has been implicated in human prostate cancer and is frequently mutated in this disease. Here, using the Nkx3.1:Cre mouse strain and a genetic deletion approach, we investigate the role of Pten specifically in the developing mouse prostate epithelia. In contrast to its role in other developing organs, this gene is dispensable for the initial developmental processes such as budding and branching. However, as cytodifferentiation progresses, abnormal luminal cells fill the ductal lumens together with augmented epithelial proliferation. This phenotype resembles the hyperplasia seen in postnatal Pten deletion models that develop neoplasia at later stages. Consistent with this, gene expression analysis showed a number of genes affected that are shared with Pten mutant prostate cancer models, including a decrease in androgen receptor regulated genes. In depth analysis of the phenotype of these mice during development revealed that loss of Pten leads to the precocious differentiation of epithelial cells towards a luminal cell fate. This study provides novel insight into the role of Pten in prostate development as part of the process of coordinating the differentiation and proliferation of cell types in time and space to form a functional organ.
Collapse
Affiliation(s)
- Isabel B. Lokody
- Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London, United Kingdom
| | - Jeffrey C. Francis
- Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London, United Kingdom
| | - Jennifer R. Gardiner
- Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London, United Kingdom
| | - Janine T. Erler
- Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London, United Kingdom
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Amanda Swain
- Division of Cancer Biology, The Institute of Cancer Research, 237 Fulham Road, London, United Kingdom
- * E-mail:
| |
Collapse
|
187
|
Trerotola M, Ganguly KK, Fazli L, Fedele C, Lu H, Dutta A, Liu Q, De Angelis T, Riddell LW, Riobo NA, Gleave ME, Zoubeidi A, Pestell RG, Altieri DC, Languino LR. Trop-2 is up-regulated in invasive prostate cancer and displaces FAK from focal contacts. Oncotarget 2015; 6:14318-28. [PMID: 26015409 PMCID: PMC4546469 DOI: 10.18632/oncotarget.3960] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 04/10/2015] [Indexed: 01/21/2023] Open
Abstract
In this study, we show that the transmembrane glycoprotein Trop-2 is up-regulated in human prostate cancer (PCa) with extracapsular extension (stages pT3/pT4) as compared to organ-confined (stage pT2) PCa. Consistent with this evidence, Trop-2 expression is found to be increased in metastatic prostate tumors of Transgenic Adenocarcinoma of Mouse Prostate mice and to strongly correlate with α5β1 integrin levels. Using PCa cells, we show that Trop-2 specifically associates with the α5 integrin subunit, as binding to α3 is not observed, and that Trop-2 displaces focal adhesion kinase from focal contacts. In support of the role of Trop-2 as a promoter of PCa metastatic phenotype, we observe high expression of this molecule in exosomes purified from Trop-2-positive PCa cells. These vesicles are then found to promote migration of Trop-2-negative PCa cells on fibronectin, an α5β1 integrin/focal adhesion kinase substrate, thus suggesting that the biological function of Trop-2 may be propagated to recipient cells. In summary, our findings show that Trop-2 promotes an α5β1 integrin-dependent pro-metastatic signaling pathway in PCa cells and that the altered expression of Trop-2 may be utilized for early identification of capsule-invading PCa.
Collapse
Affiliation(s)
- Marco Trerotola
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
- Current address: Ce.S.I. – University of Chieti-Pescara, Chieti Scalo, Italy
| | - Kirat K. Ganguly
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ladan Fazli
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Carmine Fedele
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Huimin Lu
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Anindita Dutta
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Qin Liu
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Tumor Microenvironment and Metastasis Program, The Wistar Institute Cancer Center, Philadelphia, PA, USA
| | - Tiziana De Angelis
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Luke W. Riddell
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Natalia A. Riobo
- Department of Biochemistry, Thomas Jefferson University, Philadelphia, PA, USA
| | - Martin E. Gleave
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amina Zoubeidi
- The Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Richard G. Pestell
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Dario C. Altieri
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Tumor Microenvironment and Metastasis Program, The Wistar Institute Cancer Center, Philadelphia, PA, USA
| | - Lucia R. Languino
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
188
|
Shvartsur A, Bonavida B. Trop2 and its overexpression in cancers: regulation and clinical/therapeutic implications. Genes Cancer 2015; 6:84-105. [PMID: 26000093 PMCID: PMC4426947 DOI: 10.18632/genesandcancer.40] [Citation(s) in RCA: 237] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 10/19/2014] [Indexed: 12/13/2022] Open
Abstract
Trop2 is a transmembrane glycoprotein encoded by the Tacstd2 gene. It is an intracellular calcium signal transducer that is differentially expressed in many cancers. It signals cells for self-renewal, proliferation, invasion, and survival. It has stem cell-like qualities. Trop2 is expressed in many normal tissues, though in contrast, it is overexpressed in many cancers and the overexpression of Trop2 is of prognostic significance. Several ligands have been proposed that interact with Trop2. Trop2 signals the cells via different pathways and it is transcriptionally regulated by a complex network of several transcription factors. Trop2 expression in cancer cells has been correlated with drug resistance. Several strategies target Trop2 on cancer cells that include antibodies, antibody fusion proteins, chemical inhibitors, nanoparticles, etc. The in vitro studies and pre-clinical studies, using these various therapeutic treatments, have resulted in significant inhibition of tumor cell growth both in vitro and in vivo in mice. A clinical study is underway using IMMU-132 (hrS7 linked to SN38) in patients with epithelial cancers. This review describes briefly the various characteristics of cancer cells overexpressing Trop2 and the potential application of Trop2 as both a prognostic biomarker and as a therapeutic target to reverse resistance.
Collapse
Affiliation(s)
- Anna Shvartsur
- Department of Microbiology, Immunology and Molecular Genetics, Jonsson Comprehensive Cancer Center and David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Benjamin Bonavida
- Department of Microbiology, Immunology and Molecular Genetics, Jonsson Comprehensive Cancer Center and David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
189
|
Cardillo TM, Govindan SV, Sharkey RM, Trisal P, Arrojo R, Liu D, Rossi EA, Chang CH, Goldenberg DM. Sacituzumab Govitecan (IMMU-132), an Anti-Trop-2/SN-38 Antibody-Drug Conjugate: Characterization and Efficacy in Pancreatic, Gastric, and Other Cancers. Bioconjug Chem 2015; 26:919-31. [PMID: 25915780 DOI: 10.1021/acs.bioconjchem.5b00223] [Citation(s) in RCA: 200] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Sacituzumab govitecan (IMMU-132) is an antibody-drug conjugate (ADC) made from a humanized anti-Trop-2 monoclonal antibody (hRS7) conjugated with the active metabolite of irinotecan, SN-38. In addition to its further characterization, as the clinical utility of IMMU-132 expands to an ever-widening range of Trop-2-expressing solid tumor types, its efficacy in new disease models needs to be explored in a nonclinical setting. Unlike most ADCs that use ultratoxic drugs and stable linkers, IMMU-132 uses a moderately toxic drug with a moderately stable carbonate bond between SN-38 and the linker. Flow cytometry and immunohistochemistry disclosed that Trop-2 is expressed in a wide range of tumor types, including gastric, pancreatic, triple-negative breast (TNBC), colonic, prostate, and lung. While cell-binding experiments reveal no significant differences between IMMU-132 and parental hRS7 antibody, surface plasmon resonance analysis using a Trop-2 CM5 chip shows a significant binding advantage for IMMU-132 over hRS7. The conjugate retained binding to the neonatal receptor, but it lost greater than 60% of the antibody-dependent cell-mediated cytotoxicity activity compared to that of hRS7. Exposure of tumor cells to either free SN-38 or IMMU-132 demonstrated the same signaling pathways, with pJNK1/2 and p21(WAF1/Cip1) upregulation followed by cleavage of caspases 9, 7, and 3, ultimately leading to poly-ADP-ribose polymerase cleavage and double-stranded DNA breaks. Pharmacokinetics of the intact ADC in mice reveals a mean residence time (MRT) of 15.4 h, while the carrier hRS7 antibody cleared at a similar rate as that of the unconjugated antibody (MRT ∼ 300 h). IMMU-132 treatment of mice bearing human gastric cancer xenografts (17.5 mg/kg; twice weekly × 4 weeks) resulted in significant antitumor effects compared to that of mice treated with a nonspecific control. Clinically relevant dosing schemes of IMMU-132 administered either every other week, weekly, or twice weekly in mice bearing human pancreatic or gastric cancer xenografts demonstrate similar, significant antitumor effects in both models. Current Phase I/II clinical trials ( ClinicalTrials.gov , NCT01631552) confirm anticancer activity of IMMU-132 in cancers expressing Trop-2, including gastric and pancreatic cancer patients.
Collapse
Affiliation(s)
| | | | - Robert M Sharkey
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States
| | - Preeti Trisal
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States
| | - Roberto Arrojo
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States
| | - Donglin Liu
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States
| | - Edmund A Rossi
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States.,‡IBC Pharmaceuticals, Inc., Morris Plains, New Jersey 07950, United States
| | - Chien-Hsing Chang
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States.,‡IBC Pharmaceuticals, Inc., Morris Plains, New Jersey 07950, United States
| | - David M Goldenberg
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States.,‡IBC Pharmaceuticals, Inc., Morris Plains, New Jersey 07950, United States.,§Center of Molecular Medicine and Immunology, Garden State Cancer Center, Morris Plains, New Jersey 07950, United States
| |
Collapse
|
190
|
Starodub AN, Ocean AJ, Shah MA, Guarino MJ, Picozzi VJ, Vahdat LT, Thomas SS, Govindan SV, Maliakal PP, Wegener WA, Hamburger SA, Sharkey RM, Goldenberg DM. First-in-Human Trial of a Novel Anti-Trop-2 Antibody-SN-38 Conjugate, Sacituzumab Govitecan, for the Treatment of Diverse Metastatic Solid Tumors. Clin Cancer Res 2015; 21:3870-8. [PMID: 25944802 DOI: 10.1158/1078-0432.ccr-14-3321] [Citation(s) in RCA: 249] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 04/22/2015] [Indexed: 12/12/2022]
Abstract
PURPOSE Sacituzumab govitecan (IMMU-132) is an antibody-drug conjugate (ADC) targeting Trop-2, a surface glycoprotein expressed on many epithelial tumors, for delivery of SN-38, the active metabolite of irinotecan. This phase I trial evaluated this ADC as a potential therapeutic for pretreated patients with a variety of metastatic solid cancers. EXPERIMENTAL DESIGN Sacituzumab govitecan was administered on days 1 and 8 of 21-day cycles, with cycles repeated until dose-limiting toxicity or progression. Dose escalation followed a standard 3 + 3 scheme with 4 planned dose levels and dose delay or reduction allowed. RESULTS Twenty-five patients (52-60 years old, 3 median prior chemotherapy regimens) were treated at dose levels of 8 (n = 7), 10 (n = 6), 12 (n = 9), and 18 (n = 3) mg/kg. Neutropenia was dose limiting, with 12 mg/kg the maximum tolerated dose for cycle 1, but too toxic with repeated cycles. Lower doses were acceptable for extended treatment with no treatment-related grade 4 toxicities and grade 3 toxicities limited to fatigue (n = 3), neutropenia (n = 2), diarrhea (n = 1), and leukopenia (n = 1). Using CT-based RECIST 1.1, two patients achieved partial responses (triple-negative breast cancer, colon cancer) and 16 others had stable disease as best response. Twelve patients maintained disease control with continued treatment for 16 to 36 weeks; 6 survived 15 to 20+ months. No preselection of patients based on tumor Trop-2 expression was done. CONCLUSIONS Sacituzumab govitecan had acceptable toxicity and encouraging therapeutic activity in patients with difficult-to-treat cancers. The 8 and 10 mg/kg doses were selected for phase II studies.
Collapse
Affiliation(s)
| | - Allyson J Ocean
- NewYork Presbyterian Hospital/Weill Cornell Medical Center, New York, New York
| | - Manish A Shah
- NewYork Presbyterian Hospital/Weill Cornell Medical Center, New York, New York
| | - Michael J Guarino
- Christiana Care Health System Graham Cancer Center, Newark, Delaware
| | | | - Linda T Vahdat
- NewYork Presbyterian Hospital/Weill Cornell Medical Center, New York, New York
| | | | | | | | | | | | | | | |
Collapse
|
191
|
Wanger TM, Dewitt S, Collins A, Maitland NJ, Poghosyan Z, Knäuper V. Differential regulation of TROP2 release by PKC isoforms through vesicles and ADAM17. Cell Signal 2015; 27:1325-35. [PMID: 25817572 DOI: 10.1016/j.cellsig.2015.03.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 03/04/2015] [Accepted: 03/15/2015] [Indexed: 12/29/2022]
Abstract
TROP2, a cancer cell surface protein with both pro-oncogenic and anti-oncogenic properties is cleaved by ADAM17. ADAM17 dependent cleavage requires novel PKC activity which is blocked by the ADAM10/ADAM17 inhibitor GW64 as well as by the PKC inhibitor Bim-1. Full length TROP2 release is induced by classical PKC activation and blocked by Gö6979, without affecting ADAM17 dependent TROP2 cleavage. Full length TROP2 is released in ectosomes, as inhibition of endocytosis did not prevent release. Inhibition of the atypical PKC isoform PKCζ stimulated metalloproteinase dependent N-terminal alternative TROP2 cleavage. The resulting alternative TROP2 cleavage product remains membrane associated via a disulphide bond, but is released in microvesicles with an average size of 107nm. Inhibition of endocytosis following PKCζ inhibition prevented alternative cleavage and release of TROP2, suggesting that these events require endocytic uptake and exosomal release of the corresponding microvesicles. The alternative TROP2 cleavage product was also found in PC3 cell lysates following deglycosylation, and may represent a novel biomarker in prostate cancer.
Collapse
Affiliation(s)
- Tim M Wanger
- College of Biomedical and Life Sciences, Cardiff University, Dental School, Cardiff CF14 4XY, United Kingdom
| | - Sharon Dewitt
- College of Biomedical and Life Sciences, Cardiff University, Dental School, Cardiff CF14 4XY, United Kingdom
| | - Anne Collins
- YCR Cancer Research Unit, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Norman J Maitland
- YCR Cancer Research Unit, Department of Biology, University of York, York YO10 5DD, United Kingdom
| | - Zaruhi Poghosyan
- College of Biomedical and Life Sciences, Cardiff University, Medical School, Cardiff CF14 4XN, United Kingdom
| | - Vera Knäuper
- College of Biomedical and Life Sciences, Cardiff University, Dental School, Cardiff CF14 4XY, United Kingdom
| |
Collapse
|
192
|
Wang XD, Wang Q, Chen XL, Huang JF, Yin Y, Da P, Wu H. Trop2 inhibition suppresses the proliferation and invasion of laryngeal carcinoma cells via the extracellular signal-regulated kinase/mitogen-activated protein kinase pathway. Mol Med Rep 2015; 12:865-70. [PMID: 25779928 PMCID: PMC4438912 DOI: 10.3892/mmr.2015.3485] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 01/22/2015] [Indexed: 01/18/2023] Open
Abstract
The cell surface glycoprotein Trop2 is overexpressed in various types of epithelial cancer. Laryngeal carcinoma is one of the most common types of head and neck cancer and in a previous study, the expression of Trop2 in laryngeal squamous cell carcinoma (LSCC) was identified as an independent prognostic factor. However, the biological significance of Trop2 in LSCC development remains unclear. In the current study, Trop2 protein expression in fresh LSCC tissue and paracancerous tissue was investigated using western blotting. Trop2 in the Hep2 laryngeal cell line was subsequently suppressed by transfection with small interfering RNA (siRNA). The effects of knockdown of Trop2 on cell viability, migration, invasiveness and ERK/MAPK pathway activity were investigated in the current study. The expression of Trop2 in fresh LSCC tissue was demonstrated to be significantly greater than that in paracancerous tissue. Trop2 expression was also identified to be required for proliferation, migration and invasiveness of Hep2 laryngeal carcinoma cells, as all were blocked by siRNA-mediated Trop2 inhibition. Notably, the ERK/MAPK signaling pathway and cell cycle factor, cyclin D1, were identified to be suppressed following the knockdown of Trop2 in Hep2 cells. These observations suggest that Trop2 serves an oncogenic role in LSCC and has potential as a therapeutic target.
Collapse
Affiliation(s)
- Xu-Dong Wang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Qiang Wang
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Xiao-Lin Chen
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Jian-Fei Huang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Yong Yin
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Peng Da
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226000, P.R. China
| |
Collapse
|
193
|
McDougall ARA, Tolcos M, Hooper SB, Cole TJ, Wallace MJ. Trop2: from development to disease. Dev Dyn 2015; 244:99-109. [PMID: 25523132 DOI: 10.1002/dvdy.24242] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 12/08/2014] [Accepted: 12/11/2014] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Trop2 was first discovered as a biomarker of invasive trophoblast cells. Since then most research has focused on its role in tumourigenesis because it is highly expressed in the vast majority of human tumours and animal models of cancer. It is also highly expressed in stem cells and in many organs during development. RESULTS We review the multifaceted role of Trop2 during development and tumourigenesis, including its role in regulating cell proliferation and migration, self-renewal, and maintenance of basement membrane integrity. We discuss the evolution of Trop2 and its related protein Epcam (Trop1), including their distinct roles. Mutation of Trop2 leads to gelatinous drop-like corneal dystrophy, whereas over-expression of Trop2 in human tumours promotes tumour aggressiveness and increases mortality. Although Trop2 expression is sufficient to promote tumour growth, the surprising discovery that Trop2-null mice have an increased risk of tumour development has highlighted the complexity of Trop2 signaling. Recently, studies have begun to identify the mechanisms underlying TROP2’s functions, including regulated intramembrane proteolysis or specific interactions with integrin b1 and claudin proteins. CONCLUSIONS Understanding the mechanisms underlying TROP2 signaling will clarify its role during development, aid in the development of better cancer treatments and unlock a promising new direction in regenerative medicine.
Collapse
|
194
|
Abstract
Context
Accurate classification of follicular-patterned thyroid lesions is not always an easy task on routine surgical hematoxylin-eosin–stained or cytologic fine-needle aspiration specimens. The diagnostic challenges are partially due to differential diagnostic criteria that are often subtle and subjective. In the past decades, tremendous advances have been made in molecular gene profiling of tumors and diagnostic immunohistochemistry, aiding in diagnostic accuracy and proper patient management.
Objective
To evaluate the diagnostic utility of the most commonly studied immunomarkers in the field of thyroid pathology by review of the literature, using the database of indexed articles in PubMed (US National Library of Medicine) from 1976–2013.
Data Sources
Literature review, authors' research data, and personal practice experience.
Conclusions
The appropriate use of immunohistochemistry by applying a panel of immunomarkers and using a standardized technical and interpretational method may complement the morphologic assessment and aid in the accurate classification of difficult thyroid lesions.
Collapse
Affiliation(s)
- Haiyan Liu
- From the Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania
| | - Fan Lin
- From the Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania
| |
Collapse
|
195
|
Addati T, Achille G, Centrone M, Petroni S, Popescu O, Russo S, Grammatica L, Simone G. TROP-2 expression in papillary thyroid cancer: a preliminary cyto-histological study. Cytopathology 2014; 26:303-11. [DOI: 10.1111/cyt.12196] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2014] [Indexed: 12/30/2022]
Affiliation(s)
- T. Addati
- Anatomic Pathology Unit; National Cancer Research Centre Istituto Tumori ‘Giovanni Paolo II’; Bari Italy
| | - G. Achille
- Otorhinolaryngology Unit; National Cancer Research Centre Istituto Tumori ‘Giovanni Paolo II’; Bari Italy
| | - M. Centrone
- Anatomic Pathology Unit; National Cancer Research Centre Istituto Tumori ‘Giovanni Paolo II’; Bari Italy
| | - S. Petroni
- Anatomic Pathology Unit; National Cancer Research Centre Istituto Tumori ‘Giovanni Paolo II’; Bari Italy
| | - O. Popescu
- Anatomic Pathology Unit; National Cancer Research Centre Istituto Tumori ‘Giovanni Paolo II’; Bari Italy
| | - S. Russo
- Otorhinolaryngology Unit; National Cancer Research Centre Istituto Tumori ‘Giovanni Paolo II’; Bari Italy
| | - L. Grammatica
- Otorhinolaryngology Unit; National Cancer Research Centre Istituto Tumori ‘Giovanni Paolo II’; Bari Italy
| | - G. Simone
- Anatomic Pathology Unit; National Cancer Research Centre Istituto Tumori ‘Giovanni Paolo II’; Bari Italy
| |
Collapse
|
196
|
Liu X, Li S, Yi F. Trop2 gene: a novel target for cervical cancer treatment. J Cancer Res Clin Oncol 2014; 140:1331-41. [PMID: 24816726 DOI: 10.1007/s00432-014-1696-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 04/23/2014] [Indexed: 12/20/2022]
Abstract
OBJECT Trop2 plays an important role in proliferation and invasion of tumors. Extensive research has shown that the expression level of Trop2 is closely related to the progress of cervical diseases. This study was to explore the effects of Trop2 on cell proliferation and apoptosis in cervical cancer. METHODS Trop2 was knocked down by shRNA in CaSki cells. The expression level of mRNA and protein was detected by real-time PCR and western blot, respectively. Cell proliferation was determined by CCK-8 and clone formation assay; apoptosis was measured by flow cytometry; cell cycle and apoptosis-related proteins cyclinD1, P53, bcl-2, bax, caspase 3, 8 and 9 were analyzed as well to investigate possible mechanism. RESULTS Trop2 expression was effectively repressed in CaSki cells by Trop2 shRNA, which resulted in inhibition of proliferation and colony formation, whereas apoptosis rate was significantly increased. Furthermore, in Trop2 knockdown CaSki cells, the expression of cyclinD1 and bcl-2 was significantly down-regulated, while that of P53 and bax was up-regulated accompanied by increased activities of caspase 9 and 3 but not caspase 8. CONCLUSIONS Trop2 is important in proliferation and apoptosis regulation in CaSki cells, which may become a novel target for cervical cancer treatment.
Collapse
Affiliation(s)
- Xiaoqi Liu
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, China
| | | | | |
Collapse
|
197
|
Simeone P, Trerotola M, Urbanella A, Lattanzio R, Ciavardelli D, Di Giuseppe F, Eleuterio E, Sulpizio M, Eusebi V, Pession A, Piantelli M, Alberti S. A unique four-hub protein cluster associates to glioblastoma progression. PLoS One 2014; 9:e103030. [PMID: 25050814 PMCID: PMC4106866 DOI: 10.1371/journal.pone.0103030] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 06/25/2014] [Indexed: 01/09/2023] Open
Abstract
Gliomas are the most frequent brain tumors. Among them, glioblastomas are malignant and largely resistant to available treatments. Histopathology is the gold standard for classification and grading of brain tumors. However, brain tumor heterogeneity is remarkable and histopathology procedures for glioma classification remain unsatisfactory for predicting disease course as well as response to treatment. Proteins that tightly associate with cancer differentiation and progression, can bear important prognostic information. Here, we describe the identification of protein clusters differentially expressed in high-grade versus low-grade gliomas. Tissue samples from 25 high-grade tumors, 10 low-grade tumors and 5 normal brain cortices were analyzed by 2D-PAGE and proteomic profiling by mass spectrometry. This led to identify 48 differentially expressed protein markers between tumors and normal samples. Protein clustering by multivariate analyses (PCA and PLS-DA) provided discrimination between pathological samples to an unprecedented extent, and revealed a unique network of deranged proteins. We discovered a novel glioblastoma control module centered on four major network hubs: Huntingtin, HNF4α, c-Myc and 14-3-3ζ. Immunohistochemistry, western blotting and unbiased proteome-wide meta-analysis revealed altered expression of this glioblastoma control module in human glioma samples as compared with normal controls. Moreover, the four-hub network was found to cross-talk with both p53 and EGFR pathways. In summary, the findings of this study indicate the existence of a unifying signaling module controlling glioblastoma pathogenesis and malignant progression, and suggest novel targets for development of diagnostic and therapeutic procedures.
Collapse
Affiliation(s)
- Pasquale Simeone
- Unit of Cancer Pathology, Ce.S.I., Foundation University “G. d'Annunzio,” Chieti, Italy
| | - Marco Trerotola
- Unit of Cancer Pathology, Ce.S.I., Foundation University “G. d'Annunzio,” Chieti, Italy
| | - Andrea Urbanella
- Unit of Cancer Pathology, Ce.S.I., Foundation University “G. d'Annunzio,” Chieti, Italy
| | - Rossano Lattanzio
- Unit of Cancer Pathology, Ce.S.I., Foundation University “G. d'Annunzio,” Chieti, Italy
- Department of Experimental and Clinical Sciences, School of Medicine and Health Science, University “G. d'Annunzio,” Chieti, Italy
| | - Domenico Ciavardelli
- School of Human and Social Science, University “Kore” of Enna, Enna, Italy
- Molecular Neurology Unit, Ce.S.I., University “G. d'Annunzio,” Chieti, Italy
| | - Fabrizio Di Giuseppe
- Aging Research Center, Ce.S.I., University “G. d'Annunzio” Foundation, Chieti, Italy
- Department of Experimental and Clinical Sciences, School of Medicine and Health Science, University “G. d'Annunzio,” Chieti, Italy
- StemTeCh Group, Chieti, Italy
| | - Enrica Eleuterio
- Aging Research Center, Ce.S.I., University “G. d'Annunzio” Foundation, Chieti, Italy
- Department of Experimental and Clinical Sciences, School of Medicine and Health Science, University “G. d'Annunzio,” Chieti, Italy
- StemTeCh Group, Chieti, Italy
| | - Marilisa Sulpizio
- Aging Research Center, Ce.S.I., University “G. d'Annunzio” Foundation, Chieti, Italy
- Department of Experimental and Clinical Sciences, School of Medicine and Health Science, University “G. d'Annunzio,” Chieti, Italy
- StemTeCh Group, Chieti, Italy
| | - Vincenzo Eusebi
- Department of “Tutela Salute Donna, Vita nascente, Bambino e Adolescente,” Catholic University of the Sacred Heart, Policlinico Universitario “Agostino Gemelli,” Roma, Italy
| | - Annalisa Pession
- Section of Surgical Pathology, “M. Malpighi,” Bellaria Hospital, Bologna, Italy
| | - Mauro Piantelli
- Unit of Cancer Pathology, Ce.S.I., Foundation University “G. d'Annunzio,” Chieti, Italy
- Department of Experimental and Clinical Sciences, School of Medicine and Health Science, University “G. d'Annunzio,” Chieti, Italy
| | - Saverio Alberti
- Unit of Cancer Pathology, Ce.S.I., Foundation University “G. d'Annunzio,” Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Sciences, University “G. d'Annunzio,” Chieti, Italy
| |
Collapse
|
198
|
Rossi EA, Rossi DL, Cardillo TM, Chang CH, Goldenberg DM. Redirected T-Cell Killing of Solid Cancers Targeted with an Anti-CD3/Trop-2–Bispecific Antibody Is Enhanced in Combination with Interferon-α. Mol Cancer Ther 2014; 13:2341-51. [DOI: 10.1158/1535-7163.mct-14-0345] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
199
|
Noorlag R, van der Groep P, Leusink FKJ, van Hooff SR, Frank MH, Willems SM, van Es RJJ. Nodal metastasis and survival in oral cancer: Association with protein expression of SLPI, not with LCN2, TACSTD2, or THBS2. Head Neck 2014; 37:1130-6. [PMID: 24764155 DOI: 10.1002/hed.23716] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 02/17/2014] [Accepted: 04/21/2014] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Gene expression profiling revealed a strong signature predicting lymph node metastases in oral squamous cell carcinoma (OSCC). Four of the most predictive genes are secretory leukocyte protease inhibitor (SLPI), lipocalin-2 (LCN2), thrombospondin-2 (THBS2), and tumor-associated calcium signal transducer 2 (TACSTD2). This study correlates their protein expression with lymph node metastases, overall survival (OS), and disease-specific survival (DSS). METHODS Two hundred twelve patients with OSCC were included for protein expression analysis by immunohistochemistry. RESULTS SLPI expression correlates with lymph node metastases in the whole cohort, not in a subgroup of cT1 to 2N0. SLPI expression correlates with OS (hazard ratio [HR] = 0.61) and DSS (HR = 0.47) in multivariate analysis. LCN2, THBS2, and TACSTD2 show no correlation with lymph node metastases, OS, or DSS. CONCLUSION Although SLPI expression correlates with lymph node metastases, it has no additional value in determining lymph node metastases in early oral cancer. However, it is an independent predictor for both OS and DSS and therefore a relevant prognostic biomarker in OSCC.
Collapse
Affiliation(s)
- Rob Noorlag
- Department of Oral and Maxillofacial Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Petra van der Groep
- Department of Internal Medicine and Dermatology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank K J Leusink
- Department of Oral and Maxillofacial Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sander R van Hooff
- Department of Molecular Cancer Research, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Michaël H Frank
- Department of Oral and Maxillofacial Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Stefan M Willems
- Department of Internal Medicine and Dermatology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Robert J J van Es
- Department of Oral and Maxillofacial Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
200
|
Abstract
Genome‐wide SNP analyses have identified genomic variants associated with adult human height. However, these only explain a fraction of human height variation, suggesting that significant information might have been systematically missed by SNP sequencing analysis. A candidate for such non‐SNP‐linked information is DNA methylation. Regulation by DNA methylation requires the presence of CpG islands in the promoter region of candidate genes. Seventy two of 87 (82.8%), height‐associated genes were indeed found to contain CpG islands upstream of the transcription start site (USC CpG island searcher; validation: UCSC Genome Browser), which were shown to correlate with gene regulation. Consistent with this, DNA hypermethylation modules were detected in 42 height‐associated genes, versus 1.5% of control genes (P = 8.0199e−17), as were dynamic methylation changes and gene imprinting. Epigenetic heredity thus appears to be a determinant of adult human height. Major findings in mouse models and in human genetic diseases support this model. Modulation of DNA methylation are candidate to mediate environmental influence on epigenetic traits. This may help to explain progressive height changes over multiple generations, through trans‐generational heredity of progressive DNA methylation patterns. Epigenetic heredity appears to be a determinant of adult human height. Major findings in mouse models and in human genetic diseases support this model. Modulation of DNA methylation is candidate to mediate environmental influence on epigenetic traits.
Collapse
Affiliation(s)
- Pasquale Simeone
- Unit of Cancer Pathology, Department of Neuroscience and Imaging and CeSI, University "G. d'Annunzio" Foundation, Chieti Scalo, Italy
| | - Saverio Alberti
- Unit of Cancer Pathology, Department of Neuroscience and Imaging and CeSI, University "G. d'Annunzio" Foundation, Chieti Scalo, Italy
| |
Collapse
|