1
|
Lin Y, Coppo R, Onuma K, Endo H, Kondo J, Iwabuchi S, Hashimoto S, Itatani Y, Obama K, Inoue M. Growth pattern of de novo small clusters of colorectal cancer is regulated by Notch signaling at detachment. Cancer Sci 2024; 115:3648-3659. [PMID: 39300760 PMCID: PMC11531966 DOI: 10.1111/cas.16299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/08/2024] [Accepted: 07/17/2024] [Indexed: 09/22/2024] Open
Abstract
Cancer cell clusters have a higher capacity for metastasis than single cells, suggesting cancer cell clusters have biological properties different from those of single cells. The nature of de novo cancer cell clusters that are newly formed from tumor masses is largely unknown. Herein, we generated small cell clusters from colorectal cancer organoids and tracked the growth patterns of the clusters up to four cells. Growth patterns were classified into actively growing and poorly growing spheroids (PG). Notch signaling was robustly activated in small clusters immediately after dissociation, and Notch signaling inhibition markedly increased the proportion of PG spheroids. Only a limited number of PG spheroids grew under growth-permissive conditions in vitro, but xenograft tumors derived from Notch inhibited clusters showed growth rates comparable to those of untreated spheroids. Thus, de novo clusters are composed of cells with interchangeable growth fates, which are regulated in a context-dependent manner by Notch signaling.
Collapse
Affiliation(s)
- Yi‐Kai Lin
- Department of Clinical Bio‐resource Research and DevelopmentGraduate School of Medicine, Kyoto UniversityKyotoJapan
- Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Roberto Coppo
- Department of Clinical Bio‐resource Research and DevelopmentGraduate School of Medicine, Kyoto UniversityKyotoJapan
| | - Kunishige Onuma
- Department of Clinical Bio‐resource Research and DevelopmentGraduate School of Medicine, Kyoto UniversityKyotoJapan
| | - Hiroko Endo
- Department of BiochemistryOsaka International Cancer InstituteOsakaJapan
- Present address:
Carna Biosciences Inc.HyogoJapan
| | - Jumpei Kondo
- Department of Clinical Bio‐resource Research and DevelopmentGraduate School of Medicine, Kyoto UniversityKyotoJapan
- Present address:
Division of Health Sciences, Department of Molecular Biology and Clinical InvestigationGraduate School of Medicine, Osaka UniversityOsakaJapan
| | - Sadahiro Iwabuchi
- Department of Molecular PathophysiologyInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
| | - Shinichi Hashimoto
- Department of Molecular PathophysiologyInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
| | - Yoshiro Itatani
- Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Kazutaka Obama
- Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Masahiro Inoue
- Department of Clinical Bio‐resource Research and DevelopmentGraduate School of Medicine, Kyoto UniversityKyotoJapan
| |
Collapse
|
2
|
Masuda M, Iida K, Iwabuchi S, Tanaka M, Kubota S, Uematsu H, Onuma K, Kukita Y, Kato K, Kamiura S, Nakajima A, Coppo R, Kanda M, Yoshino K, Ueda Y, Morii E, Kimura T, Kondo J, Okada-Hatakeyama M, Hashimoto S, Inoue M. Clonal Origin and Lineage Ambiguity in Mixed Neuroendocrine Carcinoma of the Uterine Cervix. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:415-429. [PMID: 38103888 DOI: 10.1016/j.ajpath.2023.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/23/2023] [Accepted: 11/20/2023] [Indexed: 12/19/2023]
Abstract
Small-cell neuroendocrine carcinoma (SCNEC) of the cervix is a rare disease characterized by a high incidence of mixed tumors with other types of cancer. The mechanism underlying this mixed phenotype is not well understood. This study established a panel of organoid lines from patients with SCNEC of the cervix and ultimately focused on one line, which retained a mixed tumor phenotype, both in vitro and in vivo. Histologically, both organoids and xenograft tumors showed distinct differentiation into either SCNEC or adenocarcinoma in some regions and ambiguous differentiation in others. Tracking single cells indicated the existence of cells with bipotential differentiation toward SCNEC and adenocarcinomas. Single-cell transcriptional analysis identified three distinct clusters: SCNEC-like, adenocarcinoma-like, and a cluster lacking specific differentiation markers. The expression of neuroendocrine markers was enriched in the SCNEC-like cluster but not exclusively. Human papillomavirus 18 E6 was enriched in the SCNEC-like cluster, which showed higher proliferation and lower levels of the p53 pathway. After treatment with anticancer drugs, the expression of adenocarcinoma markers increased, whereas that of SCNEC decreased. Using a reporter system for keratin 19 expression, changes in the differentiation of each cell were shown to be associated with the shift in differentiation induced by drug treatment. These data suggest that mixed SCNEC/cervical tumors have a clonal origin and are characterized by an ambiguous and flexible differentiation state.
Collapse
Affiliation(s)
- Masamune Masuda
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Keita Iida
- Laboratory of Cell Systems, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Sadahiro Iwabuchi
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Mie Tanaka
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Satoshi Kubota
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan; Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | - Hiroyuki Uematsu
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kunishige Onuma
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoji Kukita
- Department of Molecular and Medical Genetics, Osaka International Cancer Institute, Osaka, Japan
| | - Kikuya Kato
- Department of Molecular and Medical Genetics, Osaka International Cancer Institute, Osaka, Japan
| | - Shoji Kamiura
- Department of Gynecology, Osaka International Cancer Institute, Osaka, Japan
| | - Aya Nakajima
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan; Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Roberto Coppo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mizuki Kanda
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kiyoshi Yoshino
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yutaka Ueda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Jumpei Kondo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | | | - Shinichi Hashimoto
- Department of Molecular Pathophysiology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Masahiro Inoue
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan.
| |
Collapse
|
3
|
Coppo R, Kondo J, Onuma K, Inoue M. Tracking the growth fate of single cells and isolating slow-growing cells in human colorectal cancer organoids. STAR Protoc 2023; 4:102395. [PMID: 37384521 PMCID: PMC10511865 DOI: 10.1016/j.xpro.2023.102395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/02/2023] [Accepted: 05/31/2023] [Indexed: 07/01/2023] Open
Abstract
Patient-derived tumor organoids are three-dimensionally cultured cancer cells that enable a suitable platform for studying heterogeneity and plasticity of cancer. We present a protocol for tracking the growth fate of single cells and isolating slow-growing cells in human colorectal cancer organoids. We describe steps for organoid preparation and culturing using the cancer-tissue-originating spheroid method, maintaining cell-cell contact throughout. We then detail a single-cell-derived spheroid-forming and growth assay, confirming single-cell plating, monitoring growth over time, and isolating slow-growing cells. For complete details on the use and execution of this protocol, please refer to Coppo et al.1.
Collapse
Affiliation(s)
- Roberto Coppo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Jumpei Kondo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kunishige Onuma
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Inoue
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
4
|
Shimizu S, Kondo J, Onuma K, Coppo R, Ota K, Kamada M, Harada Y, Tanaka Y, Nakazawa MA, Tamada Y, Okuno Y, Kawada K, Obama K, Coffey RJ, Fujiwara Y, Inoue M. Inhibition of the bone morphogenetic protein pathway suppresses tumor growth through downregulation of epidermal growth factor receptor in MEK/ERK-dependent colorectal cancer. Cancer Sci 2023; 114:3636-3648. [PMID: 37357017 PMCID: PMC10475764 DOI: 10.1111/cas.15882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 05/22/2023] [Accepted: 05/27/2023] [Indexed: 06/27/2023] Open
Abstract
The bone morphogenetic protein (BMP) pathway promotes differentiation and induces apoptosis in normal colorectal epithelial cells. However, its role in colorectal cancer (CRC) is controversial, where it can act as context-dependent tumor promoter or tumor suppressor. Here we have found that CRC cells reside in a BMP-rich environment based on curation of two publicly available RNA-sequencing databases. Suppression of BMP using a specific BMP inhibitor, LDN193189, suppresses the growth of select CRC organoids. Colorectal cancer organoids treated with LDN193189 showed a decrease in epidermal growth factor receptor, which was mediated by protein degradation induced by leucine-rich repeats and immunoglobulin-like domains protein 1 (LRIG1) expression. Among 18 molecularly characterized CRC organoids, suppression of growth by BMP inhibition correlated with induction of LRIG1 gene expression. Notably, knockdown of LRIG1 in organoids diminished the growth-suppressive effect of LDN193189. Furthermore, in CRC organoids, which are susceptible to growth suppression by LDN193189, simultaneous treatment with LDN193189 and trametinib, an FDA-approved MEK inhibitor, resulted in cooperative growth inhibition both in vitro and in vivo. Taken together, the simultaneous inhibition of BMP and MEK could be a novel treatment option in CRC cases, and evaluating in vitro growth suppression and LRIG1 induction by BMP inhibition using patient-derived organoids could offer functional biomarkers for predicting potential responders to this regimen.
Collapse
Affiliation(s)
- Shota Shimizu
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of MedicineTottori University Faculty of MedicineTottoriJapan
| | - Jumpei Kondo
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
- Department of BiochemistryOsaka International Cancer InstituteOsakaJapan
- Department of Molecular Biochemistry and Clinical Investigation, Division of Health ScienceOsaka University Graduate School of MedicineOsakaJapan
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Kunishige Onuma
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
| | - Roberto Coppo
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
| | - Kasumi Ota
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
| | - Mayumi Kamada
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
| | - Yohei Harada
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
| | - Yoshihisa Tanaka
- Graduate School of Pharmaceutical SciencesKyoto UniversityKyotoJapan
- RIKEN Center for Computational Science(R‐CCS)HPC/HPC‐ and AI‐driven Drug Development Platform DivisionKobeJapan
| | - Mai Adachi Nakazawa
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
- Department of Medical Data Intelligence and Data Analysis Division, Innovation Center for Health PromotionHirosaki UniversityHirosakiJapan
| | - Yoshinori Tamada
- Department of Medical Data Intelligence and Data Analysis Division, Innovation Center for Health PromotionHirosaki UniversityHirosakiJapan
| | - Yasushi Okuno
- Graduate School of Medicine and Faculty of MedicineKyoto UniversityKyotoJapan
- RIKEN Center for Computational Science(R‐CCS)HPC/HPC‐ and AI‐driven Drug Development Platform DivisionKobeJapan
| | - Kenji Kawada
- Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Kazutaka Obama
- Department of Surgery, Graduate School of MedicineKyoto UniversityKyotoJapan
| | - Robert J. Coffey
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Yoshiyuki Fujiwara
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of MedicineTottori University Faculty of MedicineTottoriJapan
| | - Masahiro Inoue
- Department of Clinical Bioresource Research and DevelopmentKyoto University Graduate School of MedicineKyotoJapan
- Department of BiochemistryOsaka International Cancer InstituteOsakaJapan
| |
Collapse
|
5
|
Kamashev D, Shaban N, Lebedev T, Prassolov V, Suntsova M, Raevskiy M, Gaifullin N, Sekacheva M, Garazha A, Poddubskaya E, Sorokin M, Buzdin A. Human Blood Serum Can Diminish EGFR-Targeted Inhibition of Squamous Carcinoma Cell Growth through Reactivation of MAPK and EGFR Pathways. Cells 2023; 12:2022. [PMID: 37626832 PMCID: PMC10453612 DOI: 10.3390/cells12162022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Regardless of the presence or absence of specific diagnostic mutations, many cancer patients fail to respond to EGFR-targeted therapeutics, and a personalized approach is needed to identify putative (non)responders. We found previously that human peripheral blood and EGF can modulate the activities of EGFR-specific drugs on inhibiting clonogenity in model EGFR-positive A431 squamous carcinoma cells. Here, we report that human serum can dramatically abolish the cell growth rate inhibition by EGFR-specific drugs cetuximab and erlotinib. We show that this phenomenon is linked with derepression of drug-induced G1S cell cycle transition arrest. Furthermore, A431 cell growth inhibition by cetuximab, erlotinib, and EGF correlates with a decreased activity of ERK1/2 proteins. In turn, the EGF- and human serum-mediated rescue of drug-treated A431 cells restores ERK1/2 activity in functional tests. RNA sequencing revealed 1271 and 1566 differentially expressed genes (DEGs) in the presence of cetuximab and erlotinib, respectively. Erlotinib- and cetuximab-specific DEGs significantly overlapped. Interestingly, the expression of 100% and 75% of these DEGs restores to the no-drug level when EGF or a mixed human serum sample, respectively, is added along with cetuximab. In the case of erlotinib, EGF and human serum restore the expression of 39% and 83% of DEGs, respectively. We further assessed differential molecular pathway activation levels and propose that EGF/human serum-mediated A431 resistance to EGFR drugs can be largely explained by reactivation of the MAPK signaling cascade.
Collapse
Affiliation(s)
- Dmitri Kamashev
- I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia;
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (N.S.); (A.B.)
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia;
| | - Nina Shaban
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (N.S.); (A.B.)
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia;
| | - Timofey Lebedev
- Engelhardt Institute of Molecular Biology, Moscow 119991, Russia; (T.L.); (V.P.)
| | - Vladimir Prassolov
- Engelhardt Institute of Molecular Biology, Moscow 119991, Russia; (T.L.); (V.P.)
| | - Maria Suntsova
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia;
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (M.R.); (E.P.)
| | - Mikhail Raevskiy
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (M.R.); (E.P.)
| | - Nurshat Gaifullin
- Department of Pathology, Faculty of Medicine, Lomonosov Moscow State University, Moscow 119992, Russia;
| | - Marina Sekacheva
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (M.R.); (E.P.)
| | - Andrew Garazha
- Oncobox Ltd., Moscow 121205, Russia;
- Omicsway Corp., Walnut, CA 91789, USA
| | - Elena Poddubskaya
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (M.R.); (E.P.)
| | - Maksim Sorokin
- I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia;
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia;
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), 1200 Brussels, Belgium
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (N.S.); (A.B.)
- Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia;
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (M.R.); (E.P.)
- PathoBiology Group, European Organization for Research and Treatment of Cancer (EORTC), 1200 Brussels, Belgium
| |
Collapse
|
6
|
Ex vivo chemosensitivity assay using primary ovarian cancer organoids for predicting clinical response and screening effective drugs. Hum Cell 2023; 36:752-761. [PMID: 36474106 DOI: 10.1007/s13577-022-00827-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022]
Abstract
Selecting the best treatment for individual patients with cancer has attracted attention for improving clinical outcomes. Recent progress in organoid culture may lead to the development of personalized medicine. Unlike molecular-targeting drugs, there are no predictive methods for patient response to standard chemotherapies for ovarian cancer. We prepared organoids using the cancer tissue-originated spheroid (CTOS) method from 61 patients with ovarian cancer with 100% success rate. Chemosensitivity assays for paclitaxel and carboplatin were performed with 84% success rate using the primary organoids from 50 patients who received the chemotherapy. A wide range of sensitivities was observed among organoids for both drugs. All four clinically resistant organoids were resistant to both drugs in 18 cases in which clinical response information was available. Five out of 18 cases (28%) were double-resistant, the response rate of which was compatible with the clinical remission rate. Carboplatin was significantly more sensitive in serous than in clear cell subtypes (P = 0.025). We generated two lines of organoids, screened 1135 drugs, and found several drugs with better combinatory effects with carboplatin than with paclitaxel. Some drugs, including afatinib, have shown an additive effect with carboplatin. The organoid sensitivity assay did not predict the clinical outcomes, both progression free and overall survival.
Collapse
|
7
|
Harada Y, Sato A, Nakamura H, Kai K, Kitamura S, Nakamura T, Kurihara Y, Ikeda S, Sueoka E, Kimura S, Sueoka-Aragane N. Anti-cancer effect of afatinib, dual inhibitor of HER2 and EGFR, on novel mutation HER2 E401G in models of patient-derived cancer. BMC Cancer 2023; 23:77. [PMID: 36690964 PMCID: PMC9872313 DOI: 10.1186/s12885-022-10428-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/08/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Precision medicine with gene panel testing based on next-generation sequencing for patients with cancer is being used increasingly in clinical practice. HER2, which encodes the human epidermal growth factor receptor 2 (HER2), is a potentially important driver gene. However, therapeutic strategies aimed at mutations in the HER2 extracellular domain have not been clarified. We therefore investigated the effect of EGFR co-targeted therapy with HER2 on patient-derived cancer models with the HER2 extracellular domain mutation E401G, based on our previous findings that this mutation has an epidermal growth factor receptor (EGFR)-mediated activation mechanism. METHODS We generated a xenograft (PDX) and a cancer tissue-originated spheroid (CTOS) from a patient's cancer containing an amplified HER2 E401G mutation. With these platforms, we compared the efficacy of afatinib, a tyrosine kinase inhibitor having anti-HER2 and anti-EGFR activity, with two other therapeutic options: lapatinib, which has similar properties but weaker EGFR inhibition, and trastuzumab plus pertuzumab, for which evidence exists of treatment efficacy against cancers with wild-type HER2 amplification. Similar experiments were also performed with H2170, a cell line with wild-type HER2 amplification, to contrast the characteristics of these drug's efficacies against HER2 E401G. RESULTS We confirmed that PDX and CTOS retained morphological and immunohistochemical characteristics and HER2 gene profiles of the original tumor. In both PDX and CTOS, afatinib reduced tumor size more than lapatinib or trastuzumab plus pertuzumab. In addition, afatinib treatment resulted in a statistically significant reduction in HER2 copy number at the end of treatment. On the other hand, in H2170 xenografts with wild-type HER2 amplification, trastuzumab plus pertuzumab was most effective. CONCLUSIONS Afatinib, a dual inhibitor of HER2 and EGFR, showed a promising effect on cancers with amplified HER2 E401G, which have an EGFR-mediated activation mechanism. Analysis of the activation mechanisms of mutations and development of therapeutic strategies based on those mechanisms are critical in precision medicine for cancer patients.
Collapse
Affiliation(s)
- Yohei Harada
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
- Graduate School of Medicine, Kyoto University, 53 Shogoin-Kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Akemi Sato
- Department of Clinical Laboratory Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Hideaki Nakamura
- Department of Transfusion Medicine, Saga University Hospital, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Keita Kai
- Department of Pathology, Saga University Hospital, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Sho Kitamura
- Department of Pathology, Saga University Hospital, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Tomomi Nakamura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Yuki Kurihara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Sadakatsu Ikeda
- Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Eisaburo Sueoka
- Department of Clinical Laboratory Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Naoko Sueoka-Aragane
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan.
| |
Collapse
|
8
|
Liu J, Huang X, Huang L, Huang J, Liang D, Liao L, Deng Y, Zhang L, Zhang B, Tang W. Organoid: Next-Generation Modeling of Cancer Research and Drug Development. Front Oncol 2022; 11:826613. [PMID: 35155215 PMCID: PMC8831330 DOI: 10.3389/fonc.2021.826613] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/28/2021] [Indexed: 01/05/2023] Open
Abstract
Colorectal carcinoma is a highly prevalent and heterogeneous gastrointestinal malignancy. The emergence of organoid technology has provided a new direction for colorectal cancer research. As a novel-type model, organoid has significant advantages compared with conventional tumor research models, characterized with the high success rate of construction and the high matching with the original tumor. These characteristics provide new possibilities to study the mechanism of colorectal carcinogenesis and improve the treatment effects. The present literature would mainly summarize the characteristics of tumor organoids and the up-to-date technique development of patient-derived organoids (PDOs) and application in colorectal cancer.
Collapse
Affiliation(s)
- Jungang Liu
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiaoliang Huang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Lihaoyun Huang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jinlian Huang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Dingyu Liang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Lixian Liao
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yuqing Deng
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Lihua Zhang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Beibei Zhang
- Institute of Biomedical Research, Yunnan University, Kunming, China
| | - Weizhong Tang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
9
|
Zhou Z, Cong L, Cong X. Patient-Derived Organoids in Precision Medicine: Drug Screening, Organoid-on-a-Chip and Living Organoid Biobank. Front Oncol 2021; 11:762184. [PMID: 35036354 PMCID: PMC8755639 DOI: 10.3389/fonc.2021.762184] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Organoids are in vitro self-assembling, organ-like, three-dimensional cellular structures that stably retain key characteristics of the respective organs. Organoids can be generated from healthy or pathological tissues derived from patients. Cancer organoid culture platforms have several advantages, including conservation of the cellular composition that captures the heterogeneity and pharmacotypic signatures of the parental tumor. This platform has provided new opportunities to fill the gap between cancer research and clinical outcomes. Clinical trials have been performed using patient-derived organoids (PDO) as a tool for personalized medical decisions to predict patients' responses to therapeutic regimens and potentially improve treatment outcomes. Living organoid biobanks encompassing several cancer types have been established, providing a representative collection of well-characterized models that will facilitate drug development. In this review, we highlight recent developments in the generation of organoid cultures and PDO biobanks, in preclinical drug discovery, and methods to design a functional organoid-on-a-chip combined with microfluidic. In addition, we discuss the advantages as well as limitations of human organoids in patient-specific therapy and highlight possible future directions.
Collapse
Affiliation(s)
- Zilong Zhou
- Biobank, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Lele Cong
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xianling Cong
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
10
|
Luo Z, Zhou X, Mandal K, He N, Wennerberg W, Qu M, Jiang X, Sun W, Khademhosseini A. Reconstructing the tumor architecture into organoids. Adv Drug Deliv Rev 2021; 176:113839. [PMID: 34153370 PMCID: PMC8560135 DOI: 10.1016/j.addr.2021.113839] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023]
Abstract
Cancer remains a leading health burden worldwide. One of the challenges hindering cancer therapy development is the substantial discrepancies between the existing cancer models and the tumor microenvironment (TME) of human patients. Constructing tumor organoids represents an emerging approach to recapitulate the pathophysiological features of the TME in vitro. Over the past decade, various approaches have been demonstrated to engineer tumor organoids as in vitro cancer models, such as incorporating multiple cellular populations, reconstructing biophysical and chemical traits, and even recapitulating structural features. In this review, we focus on engineering approaches for building tumor organoids, including biomaterial-based, microfabrication-assisted, and synthetic biology-facilitated strategies. Furthermore, we summarize the applications of engineered tumor organoids in basic cancer research, cancer drug discovery, and personalized medicine. We also discuss the challenges and future opportunities in using tumor organoids for broader applications.
Collapse
Affiliation(s)
- Zhimin Luo
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA 90095, USA; School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xingwu Zhou
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Na He
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Wally Wennerberg
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Moyuan Qu
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, and Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Xing Jiang
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA 90095, USA; School of Nursing, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wujin Sun
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA.
| | - Ali Khademhosseini
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA; Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, Department of Radiology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
11
|
Norkin M, Ordóñez-Morán P, Huelsken J. High-content, targeted RNA-seq screening in organoids for drug discovery in colorectal cancer. Cell Rep 2021; 35:109026. [PMID: 33882314 DOI: 10.1016/j.celrep.2021.109026] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 12/22/2020] [Accepted: 03/31/2021] [Indexed: 12/12/2022] Open
Abstract
Organoids allow the recapitulation of intestinal homeostasis and cancerogenesis in vitro; however, RNA sequencing (RNA-seq)-based methods for drug screens are missing. We develop targeted organoid sequencing (TORNADO-seq), a high-throughput, high-content drug discovery platform that uses targeted RNA-seq to monitor the expression of large gene signatures for the detailed evaluation of cellular phenotypes in organoids. TORNADO-seq is a fast, highly reproducible time- and cost-effective ($5 per sample) method that can probe cell mixtures and their differentiation state in the intestinal system. We apply this method to isolate drugs that enrich for differentiated cell phenotypes and show that these drugs are highly efficacious against cancer compared to wild-type organoids. Furthermore, TORNADO-seq facilitates in-depth insight into the mode of action of these drugs. Our technology can easily be adapted to many other systems and will allow for more systematic, large-scale, and quantitative approaches to study the biology of complex cellular systems.
Collapse
Affiliation(s)
- Maxim Norkin
- Swiss Institute for Experimental Cancer Research (ISREC), École Polytechnique Fédérale de Lausanne-(EPFL-SV), 1015 Lausanne, Switzerland
| | - Paloma Ordóñez-Morán
- Department of Cancer and Stem Cells, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Joerg Huelsken
- Swiss Institute for Experimental Cancer Research (ISREC), École Polytechnique Fédérale de Lausanne-(EPFL-SV), 1015 Lausanne, Switzerland.
| |
Collapse
|
12
|
Seyfoori A, Barough MS, Amereh M, Jush BK, Lum JJ, Akbari M. Bioengineered tissue models for the development of dynamic immuno-associated tumor models and high-throughput immunotherapy cytotoxicity assays. Drug Discov Today 2020; 26:455-473. [PMID: 33253917 DOI: 10.1016/j.drudis.2020.11.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 10/27/2020] [Accepted: 11/24/2020] [Indexed: 01/02/2023]
Abstract
Cancer immunotherapy is rapidly developing, with numerous therapies approved over the past decade and more therapies expected to gain approval in the future. However, immunotherapy of solid tumors has been less successful because immunosuppressive barriers limit immune cell trafficking and function against cancer cells. Interactions between suppressive immune cells, cytokines, and inhibitory factors are central to cancer immunotherapy approaches. In this review, we discuss recent advances in utilizing microfluidic platforms for understanding cancer-suppressive immune system interactions. Dendritic cell (DC)-mediated tumor models, infiltrated lymphocyte-mediated tumor models [e.g., natural killer (NK) cells, T cells, chimeric antigen receptor (CAR) T cells, and macrophages], monocyte-mediated tumor models, and immune checkpoint blockade (ICB) tumor models are among the various bioengineered immune cell-cancer cell interactions that we reviewed herein.
Collapse
Affiliation(s)
- Amir Seyfoori
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
| | | | - Meitham Amereh
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Bardia Khun Jush
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Julian J Lum
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada; Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Mohsen Akbari
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; Center for Biomedical Research, University of Victoria, Victoria, BC V8P 5C2, Canada; Center for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, BC V8P 5C2, Canada.
| |
Collapse
|
13
|
Kubota S, Tanaka M, Endo H, Ito Y, Onuma K, Ueda Y, Kamiura S, Yoshino K, Kimura T, Kondo J, Inoue M. Dedifferentiation of neuroendocrine carcinoma of the uterine cervix in hypoxia. Biochem Biophys Res Commun 2020; 524:398-404. [PMID: 32007268 DOI: 10.1016/j.bbrc.2020.01.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 01/03/2020] [Indexed: 11/28/2022]
Abstract
Neuroendocrine carcinoma of small cell type (SCNEC) is a rare pathological subtype in cervical cancer, which has a worse prognosis than other histological cell types. Due to its low incidence and the lack of experimental platforms, the molecular characteristics of SCNEC in the cervix remain largely unknown. Using the cancer tissue-originated spheroid (CTOS) method-an ex vivo 3D culture system that preserves the differentiation status of the original tumors-we established a panel of CTOS lines of SCNEC. We demonstrated that xenograft tumors and CTOSs, respectively, exhibited substantial intra-tumor and intra-CTOS variation in the expression levels of chromogranin A (CHGA), a neuroendocrine tumor marker. Since hypoxia affects differentiation in various tumors and in stem cells, we also investigated how hypoxia affected neuroendocrine differentiation of SCNEC of the uterine cervix. In the CTOS line cerv21, hypoxia suppressed expression of the neuroendocrine markers CHGA and synaptophysin (SYP). Flow cytometry analysis using CD99 (a membrane protein marker of SCNEC) revealed decreased CD99 expression in a subset of cells under hypoxic conditions. These expression changes were attenuated by HIF-1α knockdown, and by a Notch inhibitor, suggesting that these molecules played a role in the regulation of neuroendocrine differentiation. The examined SCNEC markers were suppressed under hypoxia in multiple CTOS lines. Overall, our present results indicated that neuroendocrine differentiation in SCNEC of the uterus is a variable phenotype, and that hypoxia may be one of the factors regulating the differentiation status.
Collapse
Affiliation(s)
- Satoshi Kubota
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan; Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Mie Tanaka
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan; Department of Clinical Bio-resource Research and Development, Graduate School of Medicine Kyoto University, Kyoto, Japan; Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroko Endo
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | - Yu Ito
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan; Department of Clinical Bio-resource Research and Development, Graduate School of Medicine Kyoto University, Kyoto, Japan; Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kunishige Onuma
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine Kyoto University, Kyoto, Japan
| | - Yutaka Ueda
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shoji Kamiura
- Department of Gynecology, Osaka International Cancer Institute, Osaka, Japan
| | - Kiyoshi Yoshino
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Jumpei Kondo
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan; Department of Clinical Bio-resource Research and Development, Graduate School of Medicine Kyoto University, Kyoto, Japan
| | - Masahiro Inoue
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan; Department of Clinical Bio-resource Research and Development, Graduate School of Medicine Kyoto University, Kyoto, Japan.
| |
Collapse
|
14
|
Abugomaa A, Elbadawy M. Patient-derived organoid analysis of drug resistance in precision medicine: is there a value? EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020; 5:1-5. [DOI: 10.1080/23808993.2020.1715794] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/10/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Amira Abugomaa
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
- Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| | - Mohamed Elbadawy
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, Fuchu, Japan
- Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Toukh, Egypt
| |
Collapse
|
15
|
Tuomainen K, Al-Samadi A, Potdar S, Turunen L, Turunen M, Karhemo PR, Bergman P, Risteli M, Åström P, Tiikkaja R, Grenman R, Wennerberg K, Monni O, Salo T. Human Tumor-Derived Matrix Improves the Predictability of Head and Neck Cancer Drug Testing. Cancers (Basel) 2019; 12:cancers12010092. [PMID: 31905951 PMCID: PMC7017272 DOI: 10.3390/cancers12010092] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/11/2019] [Accepted: 12/25/2019] [Indexed: 12/13/2022] Open
Abstract
In vitro cancer drug testing carries a low predictive value. We developed the human leiomyoma–derived matrix “Myogel” to better mimic the human tumor microenvironment (TME). We hypothesized that Myogel could provide an appropriate microenvironment for cancer cells, thereby allowing more in vivo–relevant drug testing. We screened 19 anticancer compounds, targeting the epidermal growth factor receptor (EGFR), MEK, and PI3K/mTOR on 12 head and neck squamous cell carcinoma (HNSCC) cell lines cultured on plastic, mouse sarcoma–derived Matrigel (MSDM), and Myogel. We applied a high-throughput drug screening assay under five different culturing conditions: cells in two-dimensional (2D) plastic wells and on top or embedded in Matrigel or Myogel. We then compared the efficacy of the anticancer compounds to the response rates of 19 HNSCC monotherapy clinical trials. Cancer cells on top of Myogel responded less to EGFR and MEK inhibitors compared to cells cultured on plastic or Matrigel. However, we found a similar response to the PI3K/mTOR inhibitors under all culturing conditions. Cells grown on Myogel more closely resembled the response rates reported in EGFR-inhibitor monotherapy clinical trials. Our findings suggest that a human tumor matrix improves the predictability of in vitro anticancer drug testing compared to current 2D and MSDM methods.
Collapse
Affiliation(s)
- Katja Tuomainen
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (K.T.); (A.A.-S.); (M.T.)
- Translational Immunology Research Program (TRIMM), University of Helsinki, 00014 Helsinki, Finland
| | - Ahmed Al-Samadi
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (K.T.); (A.A.-S.); (M.T.)
- Translational Immunology Research Program (TRIMM), University of Helsinki, 00014 Helsinki, Finland
| | - Swapnil Potdar
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00290 Helsinki, Finland; (S.P.); (L.T.); (K.W.)
| | - Laura Turunen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00290 Helsinki, Finland; (S.P.); (L.T.); (K.W.)
| | - Minna Turunen
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (K.T.); (A.A.-S.); (M.T.)
- Translational Immunology Research Program (TRIMM), University of Helsinki, 00014 Helsinki, Finland
| | - Piia-Riitta Karhemo
- Research Programs Unit, Genome-Scale Biology Program and Medicum, Biochemistry and Developmental Biology, University of Helsinki, 00014 Helsinki, Finland; (P.-R.K.); (O.M.)
| | - Paula Bergman
- Biostatistics Consulting, Department of Public Health, University of Helsinki and Helsinki University Hospital, 00014 Helsinki, Finland;
| | - Maija Risteli
- Cancer and Translational Medicine Research Unit, University of Oulu, 90014 Oulu, Finland; (M.R.); (P.Å.); (R.T.)
| | - Pirjo Åström
- Cancer and Translational Medicine Research Unit, University of Oulu, 90014 Oulu, Finland; (M.R.); (P.Å.); (R.T.)
| | - Riia Tiikkaja
- Cancer and Translational Medicine Research Unit, University of Oulu, 90014 Oulu, Finland; (M.R.); (P.Å.); (R.T.)
| | - Reidar Grenman
- Department of Otorhinolaryngology—Head and Neck Surgery, Turku University Hospital, University of Turku, 20520 Turku, Finland;
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00290 Helsinki, Finland; (S.P.); (L.T.); (K.W.)
- Biotech Research and Innovation Center, Department of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Outi Monni
- Research Programs Unit, Genome-Scale Biology Program and Medicum, Biochemistry and Developmental Biology, University of Helsinki, 00014 Helsinki, Finland; (P.-R.K.); (O.M.)
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (K.T.); (A.A.-S.); (M.T.)
- Translational Immunology Research Program (TRIMM), University of Helsinki, 00014 Helsinki, Finland
- Cancer and Translational Medicine Research Unit, University of Oulu, 90014 Oulu, Finland; (M.R.); (P.Å.); (R.T.)
- Medical Research Center, Oulu University Hospital, 90014 Oulu, Finland
- Helsinki University Hospital, 00029 Helsinki, Finland
- Correspondence: ; Tel.: +358-40-544-1560
| |
Collapse
|
16
|
Forsythe S, Pu T, Skardal A. Using organoid models to predict chemotherapy efficacy: the future of precision oncology? EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1685868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Steven Forsythe
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Tracey Pu
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Wake Forest School of Medicine, Bowman Gray Center, Winston-Salem, NC, USA
| | - Aleksander Skardal
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
- Wake Forest School of Medicine, Bowman Gray Center, Winston-Salem, NC, USA
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
- Department of Molecular Medicine and Translational Science, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| |
Collapse
|
17
|
Martin SZ, Wagner DC, Hörner N, Horst D, Lang H, Tagscherer KE, Roth W. Ex vivo tissue slice culture system to measure drug-response rates of hepatic metastatic colorectal cancer. BMC Cancer 2019; 19:1030. [PMID: 31675944 PMCID: PMC6824140 DOI: 10.1186/s12885-019-6270-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/16/2019] [Indexed: 02/08/2023] Open
Abstract
Background The lack of predictive biomarkers or test systems contributes to high failure rates of systemic therapy in metastasized colorectal carcinoma, accounting for a still unfavorable prognosis. Here, we present an ex vivo functional assay to measure drug-response based on a tissue slice culture approach. Methods Tumor tissue slices of hepatic metastases of nine patients suffering from colorectal carcinoma were cultivated for 72 h and treated with different concentrations of the clinically relevant drugs Oxaliplatin, Cetuximab and Pembrolizumab. Easy to use, objective and automated analysis routines based on the Halo platform were developed to measure changes in proliferative activity and the morphometric make-up of the tumor. Apoptotic indices were assessed semiquantitatively. Results Untreated tumor tissue slices showed high morphological comparability with the original “in vivo”-tumor, preserving proliferation and stromal-tumor interactions. All but one patients showed a dosage dependent susceptibility to treatment with Oxaliplatin, whereas only two patients showed responses to Cetuximab and Pembrolizumab, respectively. Furthermore, we identified possible non-responders to Cetuximab therapy in absence of RAS-mutations. Conclusions This is the first time to demonstrate feasibility of the tissue slice culture approach for metastatic tissue of colorectal carcinoma. An automated readout of proliferation and tumor-morphometry allows for quantification of drug susceptibility. This strongly indicates a potential value of this technique as a patient-specific test-system of targeted therapy in metastatic colorectal cancer. Co-clinical trials are needed to customize for clinical application and to define adequate read-out cut-off values.
Collapse
Affiliation(s)
- Steve Z Martin
- Institute of Pathology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany. .,Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Campus Charité Mitte, 10117, Berlin, Germany.
| | - Daniel C Wagner
- Institute of Pathology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Nina Hörner
- Institute of Pathology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - David Horst
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Campus Charité Mitte, 10117, Berlin, Germany
| | - Hauke Lang
- Department of General Visceral and Transplantation Surgery, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Katrin E Tagscherer
- Institute of Pathology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Wilfried Roth
- Institute of Pathology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| |
Collapse
|
18
|
Effects of culture method on response to EGFR therapy in head and neck squamous cell carcinoma cells. Sci Rep 2019; 9:12480. [PMID: 31462653 PMCID: PMC6713778 DOI: 10.1038/s41598-019-48764-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 08/06/2019] [Indexed: 12/17/2022] Open
Abstract
The EGFR pathway plays a critical role in head and neck squamous cell carcinoma (HNSCC). Targeted therapies against the EGFR are utilized as a treatment for HNSCCC. However, patient response is heterogeneous and molecular biomarkers are lacking to predict patient response. Therefore, functional assays where drug response is directly evaluated in tumor cells are an interesting alternative. Previous studies have shown that experimental conditions modify the drug response observed in functional assays. Thus, in this work the influence of the culture environment on response to Cetuximab (EGFR monoclonal antibody) and AZD8055 (mTOR inhibitor) was evaluated. HNSCC UM-SCC-1 and UM-SCC-47 cells were cultured in 2D monoculture and compared with: 2D co-culture with cancer-associated fibroblasts (CAF); 3D culture in collagen hydrogels; and 3D culture in tumor spheroids. The results showed UM-SCC-1 drug response significantly changed in the different culture environments; leading to an increase in drug resistance in the CAF co-culture and the 3D spheroids. Conversely, UM-SCC-47 exhibited a more constant drug response across culture conditions. In conclusion, this work highlights the importance of culture conditions that modulate response to EGFR pathway inhibition.
Collapse
|
19
|
Kondo J, Inoue M. Application of Cancer Organoid Model for Drug Screening and Personalized Therapy. Cells 2019; 8:cells8050470. [PMID: 31108870 PMCID: PMC6562517 DOI: 10.3390/cells8050470] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 12/28/2022] Open
Abstract
Drug screening—i.e., testing the effects of a number of drugs in multiple cell lines—is used for drug discovery and development, and can also be performed to evaluate the heterogeneity of a disease entity. Notably, intertumoral heterogeneity is a large hurdle to overcome for establishing standard cancer treatment, necessitating disease models better than conventional established 2D cell lines for screening novel treatment candidates. In the present review, we outline recent progress regarding experimental cancer models having more physiological and clinical relevance for drug screening, which are important for the successful evaluation of cellular response to drugs. The review is particularly focused on drug screening using the cancer organoid model, which is emerging as a better physiological disease model than conventional established 2D cell lines. We also review the use of cancer organoids to examine intertumor and intratumor heterogeneity, and introduce the perspective of the clinical use of cancer organoids to enable precision medicine.
Collapse
Affiliation(s)
- Jumpei Kondo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine Kyoto University, Kyoto 606-8501, Japan.
| | - Masahiro Inoue
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
20
|
Lee IC. Cancer-on-a-chip for Drug Screening. Curr Pharm Des 2019; 24:5407-5418. [DOI: 10.2174/1381612825666190206235233] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/02/2019] [Indexed: 12/24/2022]
Abstract
:
The oncology pharmaceutical research spent a shocking amount of money on target validation and
drug optimization in preclinical models because many oncology drugs fail during clinical trial phase III. One of
the most important reasons for oncology drug failures in clinical trials may due to the poor predictive tool of
existing preclinical models. Therefore, in cancer research and personalized medicine field, it is critical to improve
the effectiveness of preclinical predictions of the drug response of patients to therapies and to reduce costly failures
in clinical trials. Three dimensional (3D) tumor models combine micro-manufacturing technologies mimic
critical physiologic parameters present in vivo, including complex multicellular architecture with multicellular
arrangement and extracellular matrix deposition, packed 3D structures with cell–cell interactions, such as tight
junctions, barriers to mass transport of drugs, nutrients and other factors, which are similar to in vivo tumor tissues.
These systems provide a solution to mimic the physiological environment for improving predictive accuracy
in oncology drug discovery.
:
his review gives an overview of the innovations, development and limitations of different types of tumor-like
construction techniques such as self-assemble spheroid formation, spheroids formation by micro-manufacturing
technologies, micro-dissected tumor tissues and tumor organoid. Combination of 3D tumor-like construction and
microfluidic techniques to achieve tumor on a chip for in vitro tumor environment modeling and drug screening
were all included. Eventually, developmental directions and technical challenges in the research field are also
discussed. We believe tumor on chip models have provided better sufficient clinical predictive power and will
bridge the gap between proof-of-concept studies and a wider implementation within the oncology drug development
for pathophysiological applications.
Collapse
Affiliation(s)
- I-Chi Lee
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
21
|
Manfredonia C, Muraro MG, Hirt C, Mele V, Governa V, Papadimitropoulos A, Däster S, Soysal SD, Droeser RA, Mechera R, Oertli D, Rosso R, Bolli M, Zettl A, Terracciano LM, Spagnoli GC, Martin I, Iezzi G. Maintenance of Primary Human Colorectal Cancer Microenvironment Using a Perfusion Bioreactor-Based 3D Culture System. ACTA ACUST UNITED AC 2019; 3:e1800300. [PMID: 32627426 DOI: 10.1002/adbi.201800300] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/03/2019] [Indexed: 12/15/2022]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related death. Conventional chemotherapeutic regimens have limited success rates, and a major challenge for the development of novel therapies is the lack of adequate in vitro models. Nonmalignant mesenchymal and immune cells of the tumor microenvironment (TME) are known to critically affect CRC progression and drug responsiveness. However, tumor drug sensitivity is still evaluated on systems, such as cell monolayers, spheroids, or tumor xenografts, which typically neglect the original TME. Here, it is investigated whether a bioreactor-based 3D culture system can preserve the main TME cellular components in primary CRC samples. Freshly excised CRC fragments are inserted between two collagen scaffolds in a "sandwich-like" format and cultured under static or perfused conditions up to 3 d. Perfused cultures maintain tumor tissue architecture and densities of proliferating tumor cells to significantly higher extents than static cultures. Stromal and immune cells are also preserved and fully viable, as indicated by their responsiveness to microenvironmental stimuli. Importantly, perfusion-based cultures prove suitable for testing the sensitivity of primary tumor cells to chemotherapies currently in use for CRC. Perfusion-based culture of primary CRC specimens recapitulates TME key features and may allow assessment of tumor drug response in a patient-specific context.
Collapse
Affiliation(s)
- Celeste Manfredonia
- Cancer Immunotherapy, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland.,Tissue Engineering, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland
| | - Manuele G Muraro
- Tissue Engineering, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland.,Oncology Surgery, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland
| | - Christian Hirt
- Tissue Engineering, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland.,Oncology Surgery, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland
| | - Valentina Mele
- Cancer Immunotherapy, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland
| | - Valeria Governa
- Cancer Immunotherapy, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland.,Oncology Surgery, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland
| | - Adam Papadimitropoulos
- Tissue Engineering, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland
| | - Silvio Däster
- Department of Surgery, University Hospital of Basel, Basel, 4031, Switzerland
| | - Savas D Soysal
- Department of Surgery, University Hospital of Basel, Basel, 4031, Switzerland
| | - Raoul A Droeser
- Department of Surgery, University Hospital of Basel, Basel, 4031, Switzerland
| | - Robert Mechera
- Department of Surgery, University Hospital of Basel, Basel, 4031, Switzerland
| | - Daniel Oertli
- Department of Surgery, University Hospital of Basel, Basel, 4031, Switzerland
| | - Raffaele Rosso
- Department of Surgery, Canton Hospital Lugano, Lugano, 6900, Switzerland
| | - Martin Bolli
- Department of Surgery, Claraspital, Basel, 4058, Switzerland
| | | | | | - Giulio C Spagnoli
- Oncology Surgery, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland
| | - Ivan Martin
- Tissue Engineering, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland
| | - Giandomenica Iezzi
- Cancer Immunotherapy, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, 4031, Switzerland.,Department of Surgery, Ente Ospedaliero Cantonale and Università Svizzera Italiana, Bellinzona, 6500, Switzerland
| |
Collapse
|
22
|
Kondo J, Ekawa T, Endo H, Yamazaki K, Tanaka N, Kukita Y, Okuyama H, Okami J, Imamura F, Ohue M, Kato K, Nomura T, Kohara A, Mori S, Dan S, Inoue M. High-throughput screening in colorectal cancer tissue-originated spheroids. Cancer Sci 2018; 110:345-355. [PMID: 30343529 PMCID: PMC6317944 DOI: 10.1111/cas.13843] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/12/2018] [Accepted: 10/19/2018] [Indexed: 12/13/2022] Open
Abstract
Patient‐derived cancer organoid culture is an important live material that reflects clinical heterogeneity. However, the limited amount of organoids available for each case as well as the considerable amount of time and cost to expand in vitro makes it impractical to perform high‐throughput drug screening using organoid cultures from multiple patients. Here, we report an advanced system for the high‐throughput screening of 2427 drugs using the cancer tissue‐originated spheroid (CTOS) method. In this system, we apply the CTOS method in an ex vivo platform from xenograft tumors, using machines to handle CTOS and reagents, and testing a CTOS reference panel of multiple CTOS lines for the hit drugs. CTOS passages in xenograft tumors resulted in minimal changes of morphological and genomic status, and xenograft tumor generation efficiently expanded the number of CTOS to evaluate multiple drugs. Our panel of colorectal cancer CTOS lines exhibited diverse sensitivities to the hit compounds, demonstrating the usefulness of this system for investigating highly heterogeneous disease.
Collapse
Affiliation(s)
- Jumpei Kondo
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | - Tomoya Ekawa
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | - Hiroko Endo
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | - Kanami Yamazaki
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Norio Tanaka
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Tokyo, Japan
| | - Yoji Kukita
- Department of Molecular and Medical Genetics, Osaka International Cancer Institute, Osaka, Japan
| | - Hiroaki Okuyama
- Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | - Jiro Okami
- Department of Thoracic Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Fumio Imamura
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Masayuki Ohue
- Department of Gastrointestinal Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Kikuya Kato
- Department of Molecular and Medical Genetics, Osaka International Cancer Institute, Osaka, Japan
| | - Taisei Nomura
- Animal Models of Human Diseases, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Arihiro Kohara
- Laboratory of Cell Cultures, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Seiichi Mori
- Project for Development of Innovative Research on Cancer Therapeutics, Cancer Precision Medicine Center, Tokyo, Japan
| | - Shingo Dan
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masahiro Inoue
- Department of Clinical Bio-resource Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
23
|
Nichols RJ, Haderk F, Stahlhut C, Schulze CJ, Hemmati G, Wildes D, Tzitzilonis C, Mordec K, Marquez A, Romero J, Hsieh T, Zaman A, Olivas V, McCoach C, Blakely CM, Wang Z, Kiss G, Koltun ES, Gill AL, Singh M, Goldsmith MA, Smith JAM, Bivona TG. RAS nucleotide cycling underlies the SHP2 phosphatase dependence of mutant BRAF-, NF1- and RAS-driven cancers. Nat Cell Biol 2018; 20:1064-1073. [PMID: 30104724 PMCID: PMC6115280 DOI: 10.1038/s41556-018-0169-1] [Citation(s) in RCA: 301] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 07/16/2018] [Indexed: 12/24/2022]
Abstract
Oncogenic alterations in the RAS/RAF/MEK/ERK pathway drive the growth of a wide spectrum of cancers. While BRAF and MEK inhibitors are efficacious against BRAFV600E-driven cancers, effective targeted therapies are lacking for most cancers driven by other pathway alterations, including non-V600E oncogenic BRAF, RAS GTPase-activating protein (GAP) NF1 (neurofibromin 1) loss and oncogenic KRAS. Here, we show that targeting the SHP2 phosphatase (encoded by PTPN11) with RMC-4550, a small-molecule allosteric inhibitor, is effective in human cancer models bearing RAS-GTP-dependent oncogenic BRAF (for example, class 3 BRAF mutants), NF1 loss or nucleotide-cycling oncogenic RAS (for example, KRASG12C). SHP2 inhibitor treatment decreases oncogenic RAS/RAF/MEK/ERK signalling and cancer growth by disrupting SOS1-mediated RAS-GTP loading. Our findings illuminate a critical function for SHP2 in promoting oncogenic RAS/MAPK pathway activation in cancers with RAS-GTP-dependent oncogenic BRAF, NF1 loss and nucleotide-cycling oncogenic KRAS. SHP2 inhibition is a promising molecular therapeutic strategy for patients with cancers bearing these oncogenic drivers.
Collapse
Affiliation(s)
- Robert J Nichols
- Department of Biology, Revolution Medicines, Inc., Redwood City, CA, USA
| | - Franziska Haderk
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Carlos Stahlhut
- Department of Biology, Revolution Medicines, Inc., Redwood City, CA, USA
| | | | - Golzar Hemmati
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - David Wildes
- Department of Biology, Revolution Medicines, Inc., Redwood City, CA, USA
| | | | - Kasia Mordec
- Department of Biology, Revolution Medicines, Inc., Redwood City, CA, USA
| | - Abby Marquez
- Department of Biology, Revolution Medicines, Inc., Redwood City, CA, USA
| | - Jason Romero
- Department of Biology, Revolution Medicines, Inc., Redwood City, CA, USA
| | - Tientien Hsieh
- Department of Biology, Revolution Medicines, Inc., Redwood City, CA, USA
| | - Aubhishek Zaman
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Victor Olivas
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Caroline McCoach
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Collin M Blakely
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Zhengping Wang
- Department of Development Sciences, Revolution Medicines, Inc., Redwood City, CA, USA
| | - Gert Kiss
- Department of Biology, Revolution Medicines, Inc., Redwood City, CA, USA
| | - Elena S Koltun
- Department of Chemistry, Revolution Medicines, Inc., Redwood City, CA, USA
| | - Adrian L Gill
- Department of Chemistry, Revolution Medicines, Inc., Redwood City, CA, USA
| | - Mallika Singh
- Department of Biology, Revolution Medicines, Inc., Redwood City, CA, USA
| | - Mark A Goldsmith
- Department of Biology, Revolution Medicines, Inc., Redwood City, CA, USA
- Department of Chemistry, Revolution Medicines, Inc., Redwood City, CA, USA
| | | | - Trever G Bivona
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|