1
|
Sarfraz Z, Sarfraz A, Farooq MD, Khalid M, Cheema K, Javad F, Khan T, Pervaiz Z, Sarfraz M, Jaan A, Sadiq S, Anwar J. The Current Landscape of Clinical Trials for Immunotherapy in Pancreatic Cancer: A State-of-the-Art Review. J Gastrointest Cancer 2024; 55:1026-1057. [PMID: 38976079 DOI: 10.1007/s12029-024-01078-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND Pancreatic cancer remains a lethal malignancy with a 5-year survival rate below 6% and about 500,000 deaths annually worldwide. Pancreatic adenocarcinoma, the most prevalent form, is commonly associated with diabetes, chronic pancreatitis, obesity, and smoking, mainly affecting individuals aged 60 to 80 years. This systematic review aims to evaluate the efficacy of immunotherapeutic approaches in the treatment of pancreatic cancer. METHODS A systematic search was conducted to identify clinical trials (Phases I-III) assessing immunotherapy in pancreatic cancer in PubMed/Medline, CINAHL, Scopus, and Web of Science, adhering to PRISMA Statement 2020 guidelines. The final search was completed on May 25, 2024. Ongoing trials were sourced from ClinicalTrials.gov and the World Health Organization's International Clinical Trials Registry Platform (ICTRP). Keywords such as "pancreatic," "immunotherapy," "cancer," and "clinical trial" were used across databases. Gray literature was excluded. RESULTS Phase I trials, involving 337 patients, reported a median overall survival (OS) of 13.6 months (IQR: 5-62.5 months) and a median progression-free survival (PFS) of 5.1 months (IQR: 1.9-11.7 months). Phase II/III trials pooled in a total of 1463 participants had a median OS of 12.2 months (IQR: 2.5-35.55 months) and a median PFS of 8.8 months (IQR: 1.4-33.51 months). CONCLUSIONS Immunotherapy shows potential for extending survival among pancreatic cancer patients, though results vary. The immunosuppressive nature of the tumor microenvironment and diverse patient responses underline the need for further research to optimize these therapeutic strategies.
Collapse
Affiliation(s)
- Zouina Sarfraz
- Department of Medicine, Fatima Jinnah Medical University, Queen's Road, Mozang Chungi, Lahore, Pakistan.
| | | | | | - Musfira Khalid
- Department of Medicine, Fatima Jinnah Medical University, Queen's Road, Mozang Chungi, Lahore, Pakistan
| | | | | | - Taleah Khan
- CMH Lahore Medical College and Institute of Dentistry, Lahore, Pakistan
| | - Zainab Pervaiz
- CMH Lahore Medical College and Institute of Dentistry, Lahore, Pakistan
| | | | - Ali Jaan
- Rochester General Hospital, Rochester, NY, USA
| | | | - Junaid Anwar
- Baptist Hospitals of Southeast Texas, Beaumont, TX, USA
| |
Collapse
|
2
|
Murciano-Goroff YR, Uppal M, Chen M, Harada G, Schram AM. Basket Trials: Past, Present, and Future. ANNUAL REVIEW OF CANCER BIOLOGY 2024; 8:59-80. [PMID: 38938274 PMCID: PMC11210107 DOI: 10.1146/annurev-cancerbio-061421-012927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Large-scale tumor molecular profiling has revealed that diverse cancer histologies are driven by common pathways with unifying biomarkers that can be exploited therapeutically. Disease-agnostic basket trials have been increasingly utilized to test biomarker-driven therapies across cancer types. These trials have led to drug approvals and improved the lives of patients while simultaneously advancing our understanding of cancer biology. This review focuses on the practicalities of implementing basket trials, with an emphasis on molecularly targeted trials. We examine the biologic subtleties of genomic biomarker and patient selection, discuss previous successes in drug development facilitated by basket trials, describe certain novel targets and drugs, and emphasize practical considerations for participant recruitment and study design. This review also highlights strategies for aiding patient access to basket trials. As basket trials become more common, steps to ensure equitable implementation of these studies will be critical for molecularly targeted drug development.
Collapse
Affiliation(s)
| | - Manik Uppal
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Monica Chen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Guilherme Harada
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alison M Schram
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
3
|
Rzepka D, Schenker H, Geinitz H, Silberberger E, Kaudewitz D, Schuster B, Kuhlmann L, Schonath M, Ayala Gaona H, Aschacher B, Fietkau R, Schett G, Distel L. Chromosomal radiosensitivity in oncological and non-oncological patients with rheumatoid arthritis and connective tissue diseases. Radiat Oncol 2023; 18:98. [PMID: 37287050 DOI: 10.1186/s13014-023-02291-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/27/2023] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND The risk of developing late radiotoxicity after radiotherapy in patients with high chromosomal radiosensitivity after radiotherapy could potentially be higher compared to the risk in patients with average radiosensitivity. In case of extremely high radiosensitivity, dose reduction may be appropriate. Some rheumatic diseases (RhD), including connective tissue diseases (CTDs) appear to be associated with higher radiosensitivity. The question arises as to whether patients with rheumatoid arthritis (RA) also generally have a higher radiosensitivity and whether certain parameters could indicate clues to high radiosensitivity in RA patients which would then need to be further assessed before radiotherapy. METHODS Radiosensitivity was determined in 136 oncological patients with RhD, 44 of whom were RA patients, and additionally in 34 non-oncological RA patients by three-colour fluorescence in situ hybridization (FiSH), in which lymphocyte chromosomes isolated from peripheral blood are analysed for their chromosomal aberrations of an unirradiated and an with 2 Gy irradiated blood sample. The chromosomal radiosensitivity was determined by the average number of breaks per metaphase. In addition, correlations between certain RA- or RhD-relevant disease parameters or clinical features such as the disease activity score 28 and radiosensitivity were assessed. RESULTS Some oncological patients with RhD, especially those with connective tissue diseases have significantly higher radiosensitivity compared with oncology patients without RhD. In contrast, the mean radiosensitivity of the oncological patients with RA and other RhD and the non-oncological RA did not differ. 14 of the 44 examined oncological RA-patients (31.8%) had a high radiosensitivity which is defined as ≥ 0.5 breaks per metaphase. No correlation of laboratory parameters with radiosensitivity could be established. CONCLUSIONS It would be recommended to perform radiosensitivity testing in patients with connective tissue diseases in general. We did not find a higher radiosensitivity in RA patients. In the group of RA patients with an oncological disease, a higher percentage of patients showed higher radiosensitivity, although the average radiosensitivity was not high.
Collapse
Affiliation(s)
- Dinah Rzepka
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
| | - Hannah Schenker
- Department of Internal Medicine 3 - Rheumatology and Clinical Immunology, Friedrich-Alexander- Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Hans Geinitz
- Department of Radiation Oncology, Ordensklinikum Linz Barmherzige Schwestern, Linz, Austria
| | - Elisabeth Silberberger
- Department of Radiation Oncology, Ordensklinikum Linz Barmherzige Schwestern, Linz, Austria
| | - Dorothee Kaudewitz
- Department of Haematology, Oncology and Rheumatology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Barbara Schuster
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
| | - Lukas Kuhlmann
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
| | - Miriam Schonath
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
| | - Horacio Ayala Gaona
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
| | - Bernhard Aschacher
- Department of Radiation Oncology, Ordensklinikum Linz Barmherzige Schwestern, Linz, Austria
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Clinical Immunology, Friedrich-Alexander- Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Luitpold Distel
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany.
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (CCC ER-EMN), Erlangen, Germany.
| |
Collapse
|
4
|
Dyrkheeva NS, Malakhova AA, Zakharenko AL, Okorokova LS, Shtokalo DN, Pavlova SV, Medvedev SP, Zakian SM, Nushtaeva AA, Tupikin AE, Kabilov MR, Khodyreva SN, Luzina OA, Salakhutdinov NF, Lavrik OI. Transcriptomic Analysis of CRISPR/Cas9-Mediated PARP1-Knockout Cells under the Influence of Topotecan and TDP1 Inhibitor. Int J Mol Sci 2023; 24:ijms24065148. [PMID: 36982223 PMCID: PMC10049738 DOI: 10.3390/ijms24065148] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
Topoisomerase 1 (TOP1) is an enzyme that regulates DNA topology and is essential for replication, recombination, and other processes. The normal TOP1 catalytic cycle involves the formation of a short-lived covalent complex with the 3' end of DNA (TOP1 cleavage complex, TOP1cc), which can be stabilized, resulting in cell death. This fact substantiates the effectiveness of anticancer drugs-TOP1 poisons, such as topotecan, that block the relegation of DNA and fix TOP1cc. Tyrosyl-DNA phosphodiesterase 1 (TDP1) is able to eliminate TOP1cc. Thus, TDP1 interferes with the action of topotecan. Poly(ADP-ribose) polymerase 1 (PARP1) is a key regulator of many processes in the cell, such as maintaining the integrity of the genome, regulation of the cell cycle, cell death, and others. PARP1 also controls the repair of TOP1cc. We performed a transcriptomic analysis of wild type and PARP1 knockout HEK293A cells treated with topotecan and TDP1 inhibitor OL9-119 alone and in combination. The largest number of differentially expressed genes (DEGs, about 4000 both up- and down-regulated genes) was found in knockout cells. Topotecan and OL9-119 treatment elicited significantly fewer DEGs in WT cells and negligible DEGs in PARP1-KO cells. A significant part of the changes caused by PARP1-KO affected the synthesis and processing of proteins. Differences under the action of treatment with TOP1 or TDP1 inhibitors alone were found in the signaling pathways for the development of cancer, DNA repair, and the proteasome. The drug combination resulted in DEGs in the ribosome, proteasome, spliceosome, and oxidative phosphorylation pathways.
Collapse
Affiliation(s)
- Nadezhda S Dyrkheeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | - Anastasia A Malakhova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentyeva Ave., 630090 Novosibirsk, Russia
- Federal Research Centre Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 10 Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | - Aleksandra L Zakharenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | | | - Dmitriy N Shtokalo
- AcademGene LLC, 6 Lavrentyeva Ave., 630090 Novosibirsk, Russia
- A.P. Ershov Institute of Informatics Systems SB RAS, 6 Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | - Sophia V Pavlova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentyeva Ave., 630090 Novosibirsk, Russia
- Federal Research Centre Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 10 Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | - Sergey P Medvedev
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentyeva Ave., 630090 Novosibirsk, Russia
- Federal Research Centre Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 10 Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | - Suren M Zakian
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentyeva Ave., 630090 Novosibirsk, Russia
- Federal Research Centre Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 10 Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | - Anna A Nushtaeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | - Alexey E Tupikin
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | - Marsel R Kabilov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | - Svetlana N Khodyreva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | - Olga A Luzina
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, 9 Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | - Nariman F Salakhutdinov
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, 9 Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | - Olga I Lavrik
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 8 Lavrentyeva Ave., 630090 Novosibirsk, Russia
- Department of Molecular Biology and Biotechnology, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
5
|
Zeng Y, Zhu S, Wang Z, Chen J, Dai J, Liu Z, Sun G, Liang J, Zhang X, Wang Z, Zhao J, Ni Y, Yang J, Wang M, Wei Q, Li X, Chen N, Li Z, Wang X, Shen Y, Yao J, Huang R, Liu J, Cai D, Zeng H, Shen P. Multidisciplinary Team (MDT) Discussion Improves Overall Survival Outcomes for Metastatic Renal Cell Carcinoma Patients. J Multidiscip Healthc 2023; 16:503-513. [PMID: 36865786 PMCID: PMC9971520 DOI: 10.2147/jmdh.s393457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
Purpose Multidisciplinary team (MDT) discussion is a widely used model to manage patients diagnosed with cancer. However, there has been no direct evidence to prove its effect on the prognosis of metastatic renal cell carcinoma (mRCC) patients, so this study explored the impact of MDT discussion on mRCC patient survival. Methods The clinical data of 269 mRCC patients were retrospectively collected from 2012 to 2021. The cases were grouped into the MDT and non-MDT groups, then subgroup analysis was performed according to different histology types, as well as exploring the role of MDT in patients who have undergone multiple-line therapy. Overall survival (OS) and progression free survival (PFS) were set as the study endpoint. Results Approximately half (48.0%, 129/269) of the patients were in the MDT group, with univariable survival analyses showing these patients had remarkably longer median OS (MDT group: 73.7 months; non-MDT group: 33.2 months, hazard ratio (HR): 0.423 (0.288, 0.622), p<0.001) and longer median PFS (MDT group: 16.9 months, non-MDT group: 12.7 months, HR: 0.722 (0.542, 0.962), p=0.026). Furthermore, MDT management resulted in longer survival for both ccRCC and non-ccRCC subgroups. Patients in the MDT group were more likely to receive multi-line therapy (MDT group: 79/129, 61.2% vs non-MDT group: 56/140, 40.0%, p<0.001), and within this patient group, MDT management still resulted in longer OS (MDT group: 94.0 months; non-MDT group: 43.5 months, p=0.009). Conclusion MDT is associated with prolonged overall survival in mRCC independent of histology, ensuring that patients receive better management and precise treatment.
Collapse
Affiliation(s)
- Yuhao Zeng
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Sha Zhu
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Zilin Wang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Junru Chen
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Jindong Dai
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Zhenhua Liu
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Guangxi Sun
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Jiayu Liang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Xingming Zhang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Zhipeng Wang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Jinge Zhao
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Yuchao Ni
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Jiyu Yang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Minghao Wang
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Qiang Wei
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Xiang Li
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Ni Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Zhiping Li
- Department of Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Xin Wang
- Department of Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Yali Shen
- Department of Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Jin Yao
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Rui Huang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Jiyan Liu
- Department of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Diming Cai
- Department of Medical Ultrasound, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Hao Zeng
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Pengfei Shen
- Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China,Correspondence: Pengfei Shen; Hao Zeng, Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China, Email ;
| |
Collapse
|
6
|
Martell EM, González-Garcia M, Ständker L, Otero-González AJ. Host defense peptides as immunomodulators: The other side of the coin. Peptides 2021; 146:170644. [PMID: 34464592 DOI: 10.1016/j.peptides.2021.170644] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 12/13/2022]
Abstract
Host defense peptides (HDPs) exhibit a broad range of antimicrobial and immunomodulatory activities. In this sense, both functions are like different sides of the same coin. The direct antimicrobial side was discovered first, and widely studied for the development of anti-infective therapies. In contrast, the immunomodulatory side was recognized later and in the last 20 years the interest in this field has been continuously growing. Different to their antimicrobial activities, the immunomodulatory activities of host defense peptides are more effective in vivo. They offer a great opportunity for new therapeutic applications in the fields of anti-infective therapy, chronic inflammatory diseases treatment, novel vaccine adjuvants development and anticancer immunotherapy. These immune related functions of HDPs includes chemoattraction of leukocytes, modulation of inflammation, enhancement of antigen presentation and polarization of adaptive immune responses. Our attempt with this review is to make a careful evaluation of different aspects of the less explored, but attractive immunomodulatory side of the HDP functional coin.
Collapse
Affiliation(s)
- Ernesto M Martell
- Center for Protein Studies, Faculty of Biology, Havana University, Cuba
| | | | - Ludger Ständker
- Core Facility Functional Peptidomics (CFP), Ulm University Medical Center, Ulm, Germany
| | | |
Collapse
|
7
|
Loo SY, Syn NL, Koh APF, Teng JCF, Deivasigamani A, Tan TZ, Thike AA, Vali S, Kapoor S, Wang X, Wang JW, Tan PH, Yip GW, Sethi G, Huang RYJ, Hui KM, Wang L, Goh BC, Kumar AP. Epigenetic derepression converts PPARγ into a druggable target in triple-negative and endocrine-resistant breast cancers. Cell Death Discov 2021; 7:265. [PMID: 34580286 PMCID: PMC8476547 DOI: 10.1038/s41420-021-00635-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 08/23/2021] [Accepted: 09/03/2021] [Indexed: 01/04/2023] Open
Abstract
Clinical trials repurposing peroxisome proliferator-activated receptor-gamma (PPARγ) agonists as anticancer agents have exhibited lackluster efficacy across a variety of tumor types. Here, we report that increased PPARG expression is associated with a better prognosis but is anticorrelated with histone deacetylase (HDAC) 1 and 2 expressions. We show that HDAC overexpression blunts anti-proliferative and anti-angiogenic responses to PPARγ agonists via transcriptional and post-translational mechanisms, however, these can be neutralized with clinically approved and experimental HDAC inhibitors. Supporting this notion, concomitant treatment with HDAC inhibitors was required to license the tumor-suppressive effects of PPARγ agonists in triple-negative and endocrine-refractory breast cancer cells, and combination therapy also restrained angiogenesis in a tube formation assay. This combination was also synergistic in estrogen receptor-alpha (ERα)-positive cells because HDAC blockade abrogated ERα interference with PPARγ-regulated transcription. Following a pharmacokinetics optimization study, the combination of rosiglitazone and a potent pan-HDAC inhibitor, LBH589, stalled disease progression in a mouse model of triple-negative breast cancer greater than either of the monotherapies, while exhibiting a favorable safety profile. Our findings account for historical observations of de-novo resistance to PPARγ agonist monotherapy and propound a therapeutically cogent intervention against two aggressive breast cancer subtypes.
Collapse
Affiliation(s)
- Ser Yue Loo
- Cancer Science Institute of Singapore and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Nicholas L Syn
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Angele Pei-Fern Koh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Janet Cheng-Fei Teng
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Amudha Deivasigamani
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Aye Aye Thike
- Department of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Shireen Vali
- Cellworks Research India Pvt. Ltd., Bengaluru, India
| | - Shweta Kapoor
- Cellworks Research India Pvt. Ltd., Bengaluru, India
| | - Xiaoyuan Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Cardiovascular Research Institute (CVRI), National University Heart Centre, Singapore (NUHCS), National University Health System, Singapore, Singapore
| | - Jiong Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Puay Hoon Tan
- Department of Pathology, Singapore General Hospital, Singapore, Singapore
| | - George W Yip
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ruby Yun-Ju Huang
- School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kam Man Hui
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore, Singapore
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Boon Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,National University Cancer Institute, National University Health System, Singapore, Singapore.,Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore, Singapore
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore. .,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,National University Cancer Institute, National University Health System, Singapore, Singapore.
| |
Collapse
|
8
|
Zhu S, Chen J, Ni Y, Zhang H, Liu Z, Shen P, Sun G, Liang J, Zhang X, Wang Z, Wei Q, Li X, Chen N, Li Z, Wang X, Shen Y, Yao J, Huang R, Liu J, Cai D, Zeng H. Dynamic multidisciplinary team discussions can improve the prognosis of metastatic castration-resistant prostate cancer patients. Prostate 2021; 81:721-727. [PMID: 34028061 PMCID: PMC8362088 DOI: 10.1002/pros.24167] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/18/2021] [Accepted: 05/07/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Multidisciplinary team (MDT) management is a popular treatment paradigm in managing cancer patients, which provides fully-discussed, interdisciplinary treatment recommendations for patients. However, there has been a lack of data on its actual impact on the overall survival (OS) of metastatic castration-resistant prostate cancer (mCRPC) patients. mCRPC is the end stage of prostate cancer, facing a treatment dilemma of overwhelming options; therefore, we hypothesize dynamic MDT discussions can be helpful in comprehensively managing these patients. METHODS We retrospectively collected 422 mCRPC patients' clinical information from 2013 to 2020 from our institute. Patients can voluntarily choose whether to enroll in the dynamic MDT group, which includes discussions at CRPC diagnosis and subsequent disease progression. All patients were followed up regularly, and OS from CRPC diagnosis to death was set as the endpoint of this study. RESULTS Participating in MDT discussions is a favorable independent indicator of longer overall survival (median OS: MDT (+): 39.7 months; MDT (-): 27.0 months, hazard ratio: 0.549, p = .001). Moreover, this survival benefit of MDT remained in subgroups with first-line therapy [median OS: MDT (+): not reached; MDT (-): 27.0 months, p = .001) and with multi-line therapy until the end of follow-up (median OS: MDT (+): 36.7 months; MDT (-): 25.6 months, p = .044). CONCLUSION Therefore, regular MDT discussions are valuable in the management of mCRPC patients. Clinicians are encouraged to tailor MDT discussions dynamically to provide mCRPC patients with a better and more individualized treatment plan and more prolonged survival. Take-home messages ● The MDT model is defined as dynamic MDT discussions at the time of mCRPC diagnosis and each time they progressed later on throughout the disease management. ● Prostate cancer MDT usually includes specialists in urologic oncology, pathology, chemotherapy, radiotherapy, ultrasound, imaging and nuclear medicine. ● MDT model can benefit mCRPC patients in terms of overall survival.
Collapse
Affiliation(s)
- Sha Zhu
- Department of Urology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Junru Chen
- Department of Urology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yuchao Ni
- Department of Urology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Haoran Zhang
- Department of Urology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Zhenhua Liu
- Department of Urology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Pengfei Shen
- Department of Urology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Guangxi Sun
- Department of Urology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Jiayu Liang
- Department of Urology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Xingming Zhang
- Department of Urology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Zhipeng Wang
- Department of Urology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Qiang Wei
- Department of Urology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Xiang Li
- Department of Urology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Ni Chen
- Department of Pathology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Zhiping Li
- Department of Oncology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Xin Wang
- Department of Oncology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yali Shen
- Department of Oncology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Jin Yao
- Department of Radiology, West China HospitalSichuan UniversityChengduSichuanChina
| | - Rui Huang
- Department of Nuclear Medicine, West China HospitalSichuan UniversityChengduSichuanChina
| | - Jiyan Liu
- Department of Biotherapy, West China HospitalSichuan UniversityChengduSichuanChina
| | - Diming Cai
- Department of Medical Ultrasound, West China HospitalSichuan UniversityChengduSichuanChina
| | - Hao Zeng
- Department of Urology, West China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
9
|
Krajewska M, Rauch G. A new basket trial design based on clustering of homogeneous subpopulations. J Biopharm Stat 2021; 31:425-447. [PMID: 34236938 DOI: 10.1080/10543406.2021.1897993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
As new findings in oncology suggest a focus on individualized and targeted therapies, the demand for adequate clinical trial designs rises, whereby the focus is mainly on early development phases (phase I and II). Phase II oncology trials are often planned and analysed by Simon two-stage design, which corresponds to a one-armed trial design with the option to stop early for futility. Whereas a classical phase II study focuses on one tumour type and location, the relatively new basket trial design allows testing the efficacy of a single drug simultaneously in a number of patient subsets, which correspond to different tumour types. Such trials can be analysed in various ways, including separate analyses of all baskets or by pooling across all baskets. The work presented here tries to find an adequate compromise between these two extremes by implying rules for clustering some baskets, which are reasonably homogeneous. By means of Monte-Carlo simulations, we compare the efficiency of our proposed cluster-based basket trial design with a standard approach proposed recently which only allows for complete pooling or separate analyses. The results suggest that our new design offers a considerable advantage in power, sensitivity and specificity as well as in average sample size compared to the standard approach. The proposed clustering design is an attractive option to conduct basket trials in oncology with higher efficiency and better performance.
Collapse
Affiliation(s)
- Maja Krajewska
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, Institute of Biometry and Clinical Epidemiology, Berlin, Germany
| | - Geraldine Rauch
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, Institute of Biometry and Clinical Epidemiology, Berlin, Germany
| |
Collapse
|
10
|
Goričar K, Dolžan V, Lenassi M. Extracellular Vesicles: A Novel Tool Facilitating Personalized Medicine and Pharmacogenomics in Oncology. Front Pharmacol 2021; 12:671298. [PMID: 33995103 PMCID: PMC8120271 DOI: 10.3389/fphar.2021.671298] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/19/2021] [Indexed: 01/03/2023] Open
Abstract
Biomarkers that can guide cancer therapy based on patients' individual cancer molecular signature can enable a more effective treatment with fewer adverse events. Data on actionable somatic mutations and germline genetic variants, studied by personalized medicine and pharmacogenomics, can be obtained from tumor tissue or blood samples. As tissue biopsy cannot reflect the heterogeneity of the tumor or its temporal changes, liquid biopsy is a promising alternative approach. In recent years, extracellular vesicles (EVs) have emerged as a potential source of biomarkers in liquid biopsy. EVs are a heterogeneous population of membrane bound particles, which are released from all cells and accumulate into body fluids. They contain various proteins, lipids, nucleic acids (miRNA, mRNA, and DNA) and metabolites. In cancer, EV biomolecular composition and concentration are changed. Tumor EVs can promote the remodeling of the tumor microenvironment and pre-metastatic niche formation, and contribute to transfer of oncogenic potential or drug resistance during chemotherapy. This makes them a promising source of minimally invasive biomarkers. A limited number of clinical studies investigated EVs to monitor cancer progression, tumor evolution or drug resistance and several putative EV-bound protein and RNA biomarkers were identified. This review is focused on EVs as novel biomarker source for personalized medicine and pharmacogenomics in oncology. As several pharmacogenes and genes associated with targeted therapy, chemotherapy or hormonal therapy were already detected in EVs, they might be used for fine-tuning personalized cancer treatment.
Collapse
Affiliation(s)
| | | | - Metka Lenassi
- Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
11
|
Syn NL, Chua DW, Raphael Chen L, Tan YC, Goh BKP, Chung Cheow P, Jeyaraj PR, Koh Y, Chung A, Yee Lee S, Lucien Ooi L, Tai BC, Yip Chan C, Teo JY. Time-varying prognostic effects of primary tumor sidedness and grade after curative liver resection for colorectal liver metastases. Surg Oncol 2021; 38:101586. [PMID: 33933898 DOI: 10.1016/j.suronc.2021.101586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/07/2021] [Accepted: 04/18/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND The veracity of the proportional hazards (PH) requirement is rarely scrutinized in most areas of cancer research, although fulfilment of this assumption underpins widely-used Cox survival models. We sought to critically appraise the existence of prognostic factors with time-dependent effects and to characterize their impact on survival among CLM patients. METHODS Consecutive patients who underwent liver resection with curative intent for CLM at the Singapore General Hospital were identified from a prospectively-maintained database. We evaluated PH of 55 candidate variables, and parameters which departed significantly from proportionality were included in Cox models that incorporated an interaction term to account for time-dependent effects. As sensitivity analyses, we fitted Weibull mixture 'cure' models to handle long plateaus in the tails of survival curves, and also analyzed the restricted mean survival time. RESULTS 318 consecutive patients who underwent curative liver resection for CLM between Jan 2000 and Nov 2016 were included in this analysis. Hazard ratios for tumor grade (poorly-versus well- and moderately-differentiated) were found to decrease from 3.135 (95% CI: 1.637-6.003) at 12 months to 2.048 (95% CI: 1.038-4.042) after 24 months, and ceased to be significant at 26 months. Compared to left-sided tumors, a right-sided tumor location was found to portend worse prognosis for the first 10 months after resection but subsequently confer a survival benefit due to a crossing of survival curves. Corroborating this observation, long-term cure fractions were estimated to be 25.5% (95% CI: 17.4%-33.6%) and 34.2% (95% CI: 17.4%-50.9%) among patients with left-sided and right-sided primary disease respectively. CONCLUSION Primary tumor sidedness and grade appear to exert time-varying prognostic effects in CLM patients undergoing curative liver resection.
Collapse
Affiliation(s)
- Nicholas L Syn
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Biostatistics & Modelling Domain, Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Darren W Chua
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore
| | - Lionel Raphael Chen
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore
| | - Yu Chuan Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Brian K P Goh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore; Duke-NUS Graduate Medical School, Singapore
| | - Peng Chung Cheow
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore; Duke-NUS Graduate Medical School, Singapore
| | - Prema Raj Jeyaraj
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore; Duke-NUS Graduate Medical School, Singapore
| | - Yexin Koh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore; Duke-NUS Graduate Medical School, Singapore
| | - Alexander Chung
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore; Duke-NUS Graduate Medical School, Singapore
| | - Ser Yee Lee
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore; Duke-NUS Graduate Medical School, Singapore
| | - London Lucien Ooi
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore; Duke-NUS Graduate Medical School, Singapore
| | - Bee Choo Tai
- Biostatistics & Modelling Domain, Saw Swee Hock School of Public Health, National University of Singapore, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore; Biostatistics Core, Investigational Medicine Unit, National University Health System, Singapore
| | - Chung Yip Chan
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore; Biostatistics & Modelling Domain, Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Jin Yao Teo
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore; Duke-NUS Graduate Medical School, Singapore.
| |
Collapse
|
12
|
Ubels J, Sonneveld P, van Vliet MH, de Ridder J. Gene Networks Constructed Through Simulated Treatment Learning can Predict Proteasome Inhibitor Benefit in Multiple Myeloma. Clin Cancer Res 2020; 26:5952-5961. [PMID: 32913136 DOI: 10.1158/1078-0432.ccr-20-0742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/27/2020] [Accepted: 09/03/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Proteasome inhibitors are widely used in treating multiple myeloma, but can cause serious side effects and response varies among patients. It is, therefore, important to gain more insight into which patients will benefit from proteasome inhibitors. EXPERIMENTAL DESIGN We introduce simulated treatment learned signatures (STLsig), a machine learning method to identify predictive gene expression signatures. STLsig uses genetically similar patients who have received an alternative treatment to model which patients will benefit more from proteasome inhibitors than from an alternative treatment. STLsig constructs gene networks by linking genes that are synergistic in their ability to predict benefit. RESULTS In a dataset of 910 patients with multiple myeloma, STLsig identified two gene networks that together can predict benefit to the proteasome inhibitor, bortezomib. In class "benefit," we found an HR of 0.47 (P = 0.04) in favor of bortezomib, while in class "no benefit," the HR was 0.91 (P = 0.68). Importantly, we observed a similar performance (HR class benefit, 0.46; P = 0.04) in an independent patient cohort. Moreover, this signature also predicts benefit for the proteasome inhibitor, carfilzomib, indicating it is not specific to bortezomib. No equivalent signature can be found when the genes in the signature are excluded from the analysis, indicating that they are essential. Multiple genes in the signature are linked to working mechanisms of proteasome inhibitors or multiple myeloma disease progression. CONCLUSIONS STLsig can identify gene signatures that could aid in treatment decisions for patients with multiple myeloma and provide insight into the biological mechanism behind treatment benefit.
Collapse
Affiliation(s)
- Joske Ubels
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, the Netherlands.,Oncode Institute, Utrecht, the Netherlands.,Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.,SkylineDx, Rotterdam, the Netherlands
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | - Jeroen de Ridder
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, the Netherlands. .,Oncode Institute, Utrecht, the Netherlands
| |
Collapse
|
13
|
Popova AA, Dietrich S, Huber W, Reischl M, Peravali R, Levkin PA. Miniaturized Drug Sensitivity and Resistance Test on Patient-Derived Cells Using Droplet-Microarray. SLAS Technol 2020; 26:274-286. [PMID: 32791934 DOI: 10.1177/2472630320934432] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Testing the sensitivity of patient-derived tumor cells ex vivo can potentially help determining the appropriate treatment for each patient and spot the development of resistance to a given therapy. The number of cells obtainable from a biopsy is, however, often insufficient for performing ex vivo tests in conventional microtiter plates. Here, we introduce a novel Droplet-Microarray platform based on a hydrophilic-superhydrophobic patterned surface that enables screenings using only 100 cells and 30 picomoles of a drug per individual nanoliter-sized droplet. We demonstrate that the dose-response of as few as 100 primary patient-derived chronic lymphocytic leukemia (CLL) cells to anticancer compounds on the Droplet-Microarray platform resembles the dose-response obtained in 384-well plates requiring 20,000 tumor cells per experiment. The extremely miniaturized Droplet-Microarray platform thus carries great potential for ex vivo drug sensitivity and resistance tests on patient-derived tumor cells and potentially for implementing such tests in medical practice of precision medicine.
Collapse
Affiliation(s)
- Anna A Popova
- Karlsruhe Institute of Technology, Institute of Chemical and Biological Systems, Eggenstein-Leopoldshafen, Germany
| | - Sascha Dietrich
- National Center for Tumor Diseases, Heidelberg, Germany.,Medizinische Klinik V, University Hospital of Heidelberg, Heidelberg, Germany.,European Molecular Biology Laboratories (EMBL), Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany
| | - Wolfgang Huber
- European Molecular Biology Laboratories (EMBL), Heidelberg, Germany.,Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany
| | - Markus Reischl
- Karlsruhe Institute of Technology, Institute for Automation and Applied Informatics, Eggenstein-Leopoldshafen, Germany
| | - Ravindra Peravali
- Karlsruhe Institute of Technology, Institute of Chemical and Biological Systems, Eggenstein-Leopoldshafen, Germany
| | - Pavel A Levkin
- Karlsruhe Institute of Technology, Institute of Chemical and Biological Systems, Eggenstein-Leopoldshafen, Germany.,Karlsruhe Institute of Technology, Institute of Organic Chemistry, Karlsruhe, Germany
| |
Collapse
|
14
|
Sasani N, Roghanian R, Emtiazi G, Aghaie A. A Novel Approach on Leukodepletion Filters: Investigation of Synergistic Anticancer Effect of Purified α-Defensins and Nisin. Adv Pharm Bull 2020; 11:378-384. [PMID: 33880361 PMCID: PMC8046393 DOI: 10.34172/apb.2021.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 12/09/2019] [Accepted: 04/15/2020] [Indexed: 12/22/2022] Open
Abstract
Purpose: There are number of reports available regarding defensins activity against mammalian cells besides their antimicrobial and immune regulatory activities. This study aims to investigate anticancer and apoptosis activity of the purified defensins from leukodepletion filters alone or in synergism with bacterial peptide, nisin, on prostate and colorectal cancer. Methods: Leucoflex LCR-5 filters were backflushed by an optimized elution system. Isolated granulocytes were sonicated and the supernatant treated before further purification by high performance liquid chromatography (HPLC). SDS-PAGE and western blot testing verified the fraction. Cell culture on PC-3 (human prostate adenocarcinoma), and HCT-116 (human colorectal carcinoma) were conducted following by MTT assays in addition to annexin flow cytometry for sole and synergistic effects with peptide nisin. Results: Viable and active neutrophils could recover, and α-defensins were extracted and purified. Combinations of an optimal dose of α-defensins and nisin showed a remarkable synergistic effect on cancer cell lines (over 90% and 70% for PC-3 and HCT-116, respectively). Conclusion: It also observed that less than 40% of both cells could survive after co-treatment with optimal dose. Also, apoptosis was increased after treatment by these peptides together. Annexin Vpositive populations significantly increased in percentage in comparison with control.
Collapse
Affiliation(s)
- Niloofar Sasani
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, P. O Box 81746-79441, Isfahan, Iran
| | - Rasoul Roghanian
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, P. O Box 81746-79441, Isfahan, Iran
| | - Giti Emtiazi
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, P. O Box 81746-79441, Isfahan, Iran
| | - Afsaneh Aghaie
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
15
|
Comprehensive tumor profiling-guided therapy in rare or refractory solid cancer: A feasibility study in daily clinical practice. Bull Cancer 2020; 107:410-416. [DOI: 10.1016/j.bulcan.2019.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 12/03/2019] [Accepted: 12/22/2019] [Indexed: 11/22/2022]
|
16
|
Collignon O, Gartner C, Haidich A, James Hemmings R, Hofner B, Pétavy F, Posch M, Rantell K, Roes K, Schiel A. Current Statistical Considerations and Regulatory Perspectives on the Planning of Confirmatory Basket, Umbrella, and Platform Trials. Clin Pharmacol Ther 2020; 107:1059-1067. [DOI: 10.1002/cpt.1804] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/31/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Olivier Collignon
- Competence Centre in Methodology and Statistics Luxembourg Institute of Health Strassen Luxembourg
| | - Christian Gartner
- AGES – Österreichische Agentur für Gesundheit und Ernährungssicherheit/Austrian Agency for Health and Food Safety Vienna Austria
| | - Anna‐Bettina Haidich
- Department of Hygiene Social‐Preventive Medicine & Medical Statistics Medical School Aristotle University of Thessaloniki Thessaloniki Greece
| | - Robert James Hemmings
- Consilium Hemmings Unit 96, The Maltings Business Center The Maltings Stanstead Abbotts UK
| | - Benjamin Hofner
- Paul‐Ehrlich‐Institut Federal Institute for Vaccines and Biomedicines Langen Germany
| | - Frank Pétavy
- European Medicines Agency Amsterdam The Netherlands
| | - Martin Posch
- Section for Medical Statistics Center for Medical Statistics, Informatics, and Intelligent Systems Medical University of Vienna Vienna Austria
| | - Khadija Rantell
- Medicines and Healthcare Products Regulatory Agency London UK
| | - Kit Roes
- Julius Center for Health Sciences and Primary Care University Medical Center Utrecht Utrecht The Netherlands
| | | |
Collapse
|
17
|
Popova AA, Levkin PA. Precision Medicine in Oncology: In Vitro Drug Sensitivity and Resistance Test (DSRT) for Selection of Personalized Anticancer Therapy. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900100] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Anna A. Popova
- Karlsruhe Institute of TechnologyInstitute of Toxicology and Genetics Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
| | - Pavel A. Levkin
- Karlsruhe Institute of TechnologyInstitute of Toxicology and Genetics Hermann‐von‐Helmholtz‐Platz 1 76344 Eggenstein‐Leopoldshafen Germany
- Karlsruhe Institute of TechnologyInstitute of Organic Chemistry Fritz‐Haber Weg 6 76131 Karlsruhe Germany
| |
Collapse
|
18
|
Kondrashova O, Ho GY, Au-Yeung G, Leas L, Boughtwood T, Alsop K, Zapparoli G, Dobrovic A, Ko YA, Hsu AL, Love CJ, Lunke S, Wakefield MJ, McNally O, Quinn M, Ananda S, Neesham D, Hamilton A, Grossi M, Freimund A, Kanjanapan Y, Rischin D, Traficante N, Bowtell D, Scott CL, Christie M, Taylor GR, Mileshkin L, Waring PM. Clinical Utility of Real-Time Targeted Molecular Profiling in the Clinical Management of Ovarian Cancer: The ALLOCATE Study. JCO Precis Oncol 2019; 3:1-18. [DOI: 10.1200/po.19.00019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The ALLOCATE study was designed as a pilot to demonstrate the feasibility and clinical utility of real-time targeted molecular profiling of patients with recurrent or advanced ovarian cancer for identification of potential targeted therapies. PATIENTS AND METHODS A total of 113 patients with ovarian cancer of varying histologies were recruited from two tertiary hospitals, with 99 patient cases suitable for prospective analysis. Targeted molecular and methylation profiling of fresh biopsy and archived tumor samples were performed by screening for mutations or copy-number variations in 44 genes and for promoter methylation of BRCA1 and RAD51C. RESULTS Somatic genomic or methylation events were identified in 85% of all patient cases, with potentially actionable events with defined targeted therapies (including four resistance events) detected in 60% of all patient cases. On the basis of these findings, six patients received molecularly guided therapy, three patients had unsuspected germline cancer–associated BRCA1/ 2 mutations and were referred for genetic counseling, and two intermediate differentiated (grade 2) serous ovarian carcinomas were reclassified as low grade, leading to changes in clinical management. Additionally, secondary reversion mutations in BRCA1/ 2 were identified in fresh biopsy samples of two patients, consistent with clinical platinum/poly (ADP-ribose) polymerase inhibitor resistance. Timely reporting of results if molecular testing is done at disease recurrence, as well as early referral for patients with platinum-resistant cancers, were identified as factors that could improve the clinical utility of molecular profiling. CONCLUSION ALLOCATE molecular profiling identified known genomic and methylation alterations of the different ovarian cancer subtypes and was deemed feasible and useful in routine clinical practice. Better patient selection and access to a wider range of targeted therapies or clinical trials will further enhance the clinical utility of molecular profiling.
Collapse
Affiliation(s)
- Olga Kondrashova
- University of Melbourne, Melbourne, Victoria, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Gwo-Yaw Ho
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Royal Women’s Hospital, Parkville, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - George Au-Yeung
- University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Leakhena Leas
- University of Melbourne, Melbourne, Victoria, Australia
| | | | - Kathryn Alsop
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Giada Zapparoli
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- La Trobe University, Bundoora, Victoria, Australia
| | - Alexander Dobrovic
- University of Melbourne, Melbourne, Victoria, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- La Trobe University, Bundoora, Victoria, Australia
| | - Yi-An Ko
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Arthur L. Hsu
- University of Melbourne, Melbourne, Victoria, Australia
| | - Clare J. Love
- University of Melbourne, Melbourne, Victoria, Australia
| | | | - Matthew J. Wakefield
- University of Melbourne, Melbourne, Victoria, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Orla McNally
- University of Melbourne, Melbourne, Victoria, Australia
- Royal Women’s Hospital, Parkville, Victoria, Australia
| | - Michael Quinn
- Royal Women’s Hospital, Parkville, Victoria, Australia
| | - Sumitra Ananda
- University of Melbourne, Melbourne, Victoria, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Royal Women’s Hospital, Parkville, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Anne Hamilton
- University of Melbourne, Melbourne, Victoria, Australia
- Royal Women’s Hospital, Parkville, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Marisa Grossi
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Alison Freimund
- University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Yada Kanjanapan
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Danny Rischin
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - David Bowtell
- University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Clare L. Scott
- University of Melbourne, Melbourne, Victoria, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Royal Women’s Hospital, Parkville, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Michael Christie
- University of Melbourne, Melbourne, Victoria, Australia
- Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Graham R. Taylor
- University of Melbourne, Melbourne, Victoria, Australia
- King’s College London, London, United Kingdom
| | - Linda Mileshkin
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | | |
Collapse
|
19
|
De Palma G, Di Lorenzo VF, Krol S, Paradiso AV. Urinary exosomal shuttle RNA: Promising cancer diagnosis biomarkers of lower urinary tract. Int J Biol Markers 2019; 34:101-107. [PMID: 30862241 DOI: 10.1177/1724600819827023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Prostate and bladder cancers continue to be the first and fourth most common cancers in men worldwide; thus there is an urgent need for more accurate biomarkers that can detect these types of cancer in a non-invasive way. Liquid biopsy is a new non-invasive tool for diagnosis and with a virtually unlimited supply urine is even more attractive resource since urinary exosomes have been discovered to contain RNAs that are hallmarks of cancer. It is challenging to assay those secreting lower amounts of molecules. METHODS This review, based on articles identified through a PubMed/MEDLINE search, comprehensively summarizes state of the art approaches used in the discovery and validation of exosomal RNA biomarkers purified from the urine for lower urinary tract cancer. RESULTS The combination of PCA3 and ERG has shown a relatively good improvement in diagnostic performance; examples of other potential biomarkers and the methods utilized in their discovery are also discussed in this review. CONCLUSIONS Of these last markers, to date there are still few data to implement these for routine diagnosis.
Collapse
Affiliation(s)
- Giuseppe De Palma
- 1 Institutional BioBank, Experimental Oncology and Biobank Management Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italia
| | | | - Silke Krol
- 3 Translational Nanotechnology Laboratory, Experimental Oncology and Biobank Management Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italia
| | - Angelo Virgilio Paradiso
- 1 Institutional BioBank, Experimental Oncology and Biobank Management Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italia
| |
Collapse
|
20
|
Zanders ED, Svensson F, Bailey DS. Therapy for glioblastoma: is it working? Drug Discov Today 2019; 24:1193-1201. [PMID: 30878561 DOI: 10.1016/j.drudis.2019.03.008] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 02/06/2019] [Accepted: 03/08/2019] [Indexed: 12/21/2022]
Abstract
Glioblastoma (GBM) remains one of the most intransigent of cancers, with a median overall survival of only 15 months after diagnosis. Drug treatments have largely proven ineffective; it is thought that this is related to the heterogeneous nature and plasticity of GBM-initiating stem cell lineages. Although many combination drug therapies are being positioned to address tumour heterogeneity, the most promising therapeutic approaches for GBM to date appear to be those targeting GBM by vaccination or antibody- and cell-based immunotherapy. We review the most recent clinical trials for GBM and discuss the role of adaptive clinical trials in developing personalised treatment strategies to address intra- and inter-tumoral heterogeneity.
Collapse
Affiliation(s)
- Edward D Zanders
- IOTA Pharmaceuticals Ltd, St John's Innovation Centre, Cowley Road, Cambridge CB4 0WS, UK
| | - Fredrik Svensson
- IOTA Pharmaceuticals Ltd, St John's Innovation Centre, Cowley Road, Cambridge CB4 0WS, UK
| | - David S Bailey
- IOTA Pharmaceuticals Ltd, St John's Innovation Centre, Cowley Road, Cambridge CB4 0WS, UK.
| |
Collapse
|
21
|
Wang S, Wu H, Chen F, Zhang Y, Zhang Y, Sun B. The antitumor activity of 4,4′-bipyridinium amphiphiles. RSC Adv 2019; 9:33023-33028. [PMID: 35529125 PMCID: PMC9073189 DOI: 10.1039/c9ra06172j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 09/26/2019] [Indexed: 01/10/2023] Open
Abstract
The cell growth inhibition and apoptosis induction of 4,4′-bipyridinium amphiphiles.
Collapse
Affiliation(s)
- Senlin Wang
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- PR China
| | - Hongshuai Wu
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- PR China
| | - Fanghui Chen
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- PR China
| | - Yu Zhang
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- PR China
| | - Yuchen Zhang
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- PR China
| | - Baiwang Sun
- School of Chemistry and Chemical Engineering
- Southeast University
- Nanjing 211189
- PR China
| |
Collapse
|
22
|
Herlemann A, Washington SL, Cooperberg MR. Health Care Delivery for Metastatic Hormone-sensitive Prostate Cancer Across the Globe. Eur Urol Focus 2018; 5:155-158. [PMID: 30587443 DOI: 10.1016/j.euf.2018.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/21/2018] [Accepted: 12/10/2018] [Indexed: 12/22/2022]
Abstract
Prostate cancer remains a leading cause of cancer-related death in men. Concurrently, the incidence of metastatic hormone-sensitive prostate cancer (mHSPC) at diagnosis has significantly risen as a result, in part, of recent advances in imaging. Given the increased utilization of prostate-specific membrane antigen-targeted positron emission tomography imaging and other modalities with improved accuracy in the detection of cancer, combined with changes in screening and other secular trends, more men get diagnosed at an oligometastatic stage in which timely treatment may improve survival. However, the optimal timing of initiation and the specific sequence of systemic agents are not yet clearly defined. Worldwide, both urologists and oncologists may primarily direct the medical management of mHSPC. This collaboration potentially invites differing treatment recommendations dependent upon the treating physician's medical specialty. Ideally, a shared decision-making approach incorporating multidisciplinary tumor board discussions and personalized analysis will provide personalized treatment recommendations to optimize the benefit for mHSPC patients. Here, we conducted a concise review and evaluation of existing literature, and provide one perspective on health care delivery for mHSPC worldwide. PATIENT SUMMARY: Given the improvement in imaging techniques and changes in screening practices, the incidence of metastatic hormone-sensitive prostate cancer will likely continue to rise. An early, multimodal treatment approach involving a multidisciplinary team is critical to delivering the best care to this patient population.
Collapse
Affiliation(s)
- Annika Herlemann
- Department of Urology, University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA; Department of Urology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Samuel L Washington
- Department of Urology, University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Matthew R Cooperberg
- Department of Urology, University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA.
| |
Collapse
|
23
|
Komarova AO, Drenichev MS, Dyrkheeva NS, Kulikova IV, Oslovsky VE, Zakharova OD, Zakharenko AL, Mikhailov SN, Lavrik OI. Novel group of tyrosyl-DNA-phosphodiesterase 1 inhibitors based on disaccharide nucleosides as drug prototypes for anti-cancer therapy. J Enzyme Inhib Med Chem 2018; 33:1415-1429. [PMID: 30191738 PMCID: PMC6136360 DOI: 10.1080/14756366.2018.1509210] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 07/06/2018] [Accepted: 08/04/2018] [Indexed: 02/03/2023] Open
Abstract
A new class of tyrosyl-DNA phosphodiesterase 1 (TDP1) inhibitors based on disaccharide nucleosides was identified. TDP1 plays an essential role in the resistance of cancer cells to currently used antitumour drugs based on Top1 inhibitors such as topotecan and irinotecan. The most effective inhibitors investigated in this study have IC50 values (half-maximal inhibitory concentration) in 0.4-18.5 µM range and demonstrate relatively low own cytotoxicity along with significant synergistic effect in combination with anti-cancer drug topotecan. Moreover, kinetic parameters of the enzymatic reaction and fluorescence anisotropy were measured using different types of DNA-biosensors to give a sufficient insight into the mechanism of inhibitor's action.
Collapse
Affiliation(s)
- Anastasia O. Komarova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russian Federation
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russian Federation
| | - Mikhail S. Drenichev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russian Federation
| | - Nadezhda S. Dyrkheeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Irina V. Kulikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russian Federation
| | - Vladimir E. Oslovsky
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russian Federation
| | - Olga D. Zakharova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Alexandra L. Zakharenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Sergey N. Mikhailov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russian Federation
| | - Olga I. Lavrik
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russian Federation
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russian Federation
| |
Collapse
|
24
|
Madhavan S, Subramaniam S, Brown TD, Chen JL. Art and Challenges of Precision Medicine: Interpreting and Integrating Genomic Data Into Clinical Practice. Am Soc Clin Oncol Educ Book 2018; 38:546-553. [PMID: 30231369 DOI: 10.1200/edbk_200759] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Precision medicine is at the forefront of innovation in cancer care. With the development of technologies to rapidly sequence DNA from tumors, cell-free DNA, proteins, and even metabolites coupled with the rapid decline in the cost of genomic sequencing, there has been an exponential increase in the amount of data generated for each patient diagnosed with cancer. The ability to harness this explosion of data will be critical to improving treatments for patients. Precision medicine lends itself to big data or "informatics" approaches and is focused on storing, accessing, sharing, and studying these data while taking necessary precautions to protect patients' privacy. Major cancer care stakeholders have developed a variety of systems to incorporate precision medicine technologies into patient care as soon as possible and also to provide the ability to store and analyze the omics and clinical data aggregately in the future. Scaling these precision medicine programs within the confines of health care system silos is challenging, and research consortiums are being formed to overcome these limitations. Incorporating and interpreting the results of precision medicine sequencing is complex and rapidly changing, necessitating reliance on a group of experts. This is often performed at molecular tumor boards at large academic and research institutions with available in-house expertise, but alternative models clinical decision support software or of virtual tumor boards potentially expand these advances to almost any patient, regardless of site of care. The promises of precision medicine will be more quickly realized by expanding collaborations to rapidly process and interpret the growing volumes of omics data.
Collapse
Affiliation(s)
- Subha Madhavan
- From the Innovation Center for Biomedical Informatics, Georgetown University, Washington, DC; Swedish Cancer Institute, Seattle, WA; The Ohio State University, Columbus, OH
| | - Somasundaram Subramaniam
- From the Innovation Center for Biomedical Informatics, Georgetown University, Washington, DC; Swedish Cancer Institute, Seattle, WA; The Ohio State University, Columbus, OH
| | - Thomas D Brown
- From the Innovation Center for Biomedical Informatics, Georgetown University, Washington, DC; Swedish Cancer Institute, Seattle, WA; The Ohio State University, Columbus, OH
| | - James L Chen
- From the Innovation Center for Biomedical Informatics, Georgetown University, Washington, DC; Swedish Cancer Institute, Seattle, WA; The Ohio State University, Columbus, OH
| |
Collapse
|
25
|
Role of tumor-derived exosomes in cancer metastasis. Biochim Biophys Acta Rev Cancer 2018; 1871:12-19. [PMID: 30419312 DOI: 10.1016/j.bbcan.2018.10.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/22/2018] [Accepted: 10/22/2018] [Indexed: 12/21/2022]
Abstract
The highlights of cancer research include the discovery of exosomes, which are small (30-100 nm) sized vesicular nanoparticles released virtually by all cells. Tumor-derived exosomes (TDEs) are notoriously known for orchestrating the invasion-metastasis cascade via systemic pathways that we have previously proposed (1), resulting in a paradigm shift of our understanding about the pathobiology of metastases. In principle, exosomes serve as transport medium for proteins, mRNAs and miRNAs to transmit targeted cues from the primary cell to distant sites via horizontal transfer or cell-receptor interaction. In this chapter, we seek to explore in-depth the mechanisms engendering TDE in the metastatic cascade, along with experimental models to augment our understanding. The aforementioned has also paved way for parallel advancements in the therapeutic armamentarium, as evident from pronounced efforts to exploit the metastatic process for therapeutic targeting. In this light, we aim to examine potential anti-metastatic therapeutic opportunities derived from exosomal research. Lastly, exosomes may play a crucial role in the contemporary era of "liquid biopsies", given the array of molecular information with diagnostic and predictive indications. We thus intend to end this chapter off by exploring future applications of exosomes that could illuminate shortcomings and propel advancements in biomarker research.
Collapse
|
26
|
Real-Time Tumor Gene Expression Profiling to Direct Gastric Cancer Chemotherapy: Proof-of-Concept “3G” Trial. Clin Cancer Res 2018; 24:5272-5281. [DOI: 10.1158/1078-0432.ccr-18-0193] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/28/2018] [Accepted: 07/12/2018] [Indexed: 11/16/2022]
|
27
|
Alekseyev YO, Fazeli R, Yang S, Basran R, Maher T, Miller NS, Remick D. A Next-Generation Sequencing Primer-How Does It Work and What Can It Do? Acad Pathol 2018; 5:2374289518766521. [PMID: 29761157 PMCID: PMC5944141 DOI: 10.1177/2374289518766521] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 02/14/2018] [Accepted: 02/16/2018] [Indexed: 12/28/2022] Open
Abstract
Next-generation sequencing refers to a high-throughput technology that determines the nucleic acid sequences and identifies variants in a sample. The technology has been introduced into clinical laboratory testing and produces test results for precision medicine. Since next-generation sequencing is relatively new, graduate students, medical students, pathology residents, and other physicians may benefit from a primer to provide a foundation about basic next-generation sequencing methods and applications, as well as specific examples where it has had diagnostic and prognostic utility. Next-generation sequencing technology grew out of advances in multiple fields to produce a sophisticated laboratory test with tremendous potential. Next-generation sequencing may be used in the clinical setting to look for specific genetic alterations in patients with cancer, diagnose inherited conditions such as cystic fibrosis, and detect and profile microbial organisms. This primer will review DNA sequencing technology, the commercialization of next-generation sequencing, and clinical uses of next-generation sequencing. Specific applications where next-generation sequencing has demonstrated utility in oncology are provided.
Collapse
Affiliation(s)
- Yuriy O Alekseyev
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Roghayeh Fazeli
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Shi Yang
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Raveen Basran
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Thomas Maher
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Nancy S Miller
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| | - Daniel Remick
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, USA
| |
Collapse
|
28
|
Syn NLX, Roudi R, Wang LZ, Wang L, Loh M, Huang Y, Ou SHI, Soong R, Drilon A, Wee I. Immune checkpoint inhibitors plus chemotherapy versus chemotherapy or immunotherapy for first-line treatment of advanced non-small cell lung cancer: a generic protocol. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2018. [DOI: 10.1002/14651858.cd013009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Nicholas LX Syn
- National University Cancer Institute; Department of Haematology-Oncology; 1E Kent Ridge Road NUHS Tower Block, Level 7 Singapore Singapore 119228
| | - Raheleh Roudi
- Iran University of Medical Sciences; Oncopathology Research Center; Hemmat Street, Next to Milad Tower Teheran Iran 14496-14530
| | - Louis Zizhao Wang
- National University of Singapore; Yong Loo Lin School of Medicine; Singapore Singapore
| | - Lingzhi Wang
- Cancer Science Institute of Singapore; Singapore Singapore
| | - Marie Loh
- Agency for Science, Technology and Research; Translational Laboratory in Genetic Medicine; 8A Biomedical Grove, Immunos Level 5 Singapore Singapore 138648
| | - Yiqing Huang
- National University Cancer Institute; Department of Haematology-Oncology; 1E Kent Ridge Road NUHS Tower Block, Level 7 Singapore Singapore 119228
| | - Sai-Hong Ignatius Ou
- Chao Family Comprehensive Cancer Center, University of California Irvine, Medical Center; Division of Hematology-Oncology; 101 city drive, Bldg 56, RT81, Rm 241, Orange County Irvine USA CA 92868-3298
| | - Richie Soong
- National University of Singapore; Department of Pathology; 14 Medical Drive, #12-01 Singapore Singapore 117599
| | - Alexander Drilon
- Memorial Sloan-Kettering Cancer Center; Early Drug Development Service; 1275 York Avenue New York USA NY 10065
| | - Ian Wee
- National University Cancer Institute; Department of Haematology-Oncology; 1E Kent Ridge Road NUHS Tower Block, Level 7 Singapore Singapore 119228
| |
Collapse
|
29
|
Molecular epidemiology of lung cancer in Iran: implications for drug development and cancer prevention. J Hum Genet 2018; 63:783-794. [DOI: 10.1038/s10038-018-0450-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/20/2018] [Accepted: 03/15/2018] [Indexed: 12/18/2022]
|
30
|
Syn NL, Lim PL, Kong LR, Wang L, Wong ALA, Lim CM, Loh TKS, Siemeister G, Goh BC, Hsieh WS. Pan-CDK inhibition augments cisplatin lethality in nasopharyngeal carcinoma cell lines and xenograft models. Signal Transduct Target Ther 2018; 3:9. [PMID: 29666673 PMCID: PMC5897350 DOI: 10.1038/s41392-018-0010-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 01/03/2018] [Accepted: 01/25/2018] [Indexed: 01/28/2023] Open
Abstract
In addition to their canonical roles in regulating cell cycle transition and transcription, cyclin-dependent kinases (CDKs) have been shown to coordinate DNA damage response pathways, suggesting a rational pairing of CDK inhibitors with genotoxic chemotherapeutic agents in the treatment of human malignancies. Here, we report that roniciclib (BAY1000394), a potent pan-CDK inhibitor, displays promising anti-neoplastic activity as a single agent and potentiates cisplatin lethality in preclinical nasopharyngeal carcinoma (NPC) models. Proliferation of the NPC cell lines HONE-1, CNE-2, C666-1, and HK-1 was effectively curbed by roniciclib treatment, with IC50 values between 11 and 38 nmol/L. These anticancer effects were mediated by pleiotropic mechanisms consistent with successful blockade of cell cycle CDKs 1, 2, 3, and 4 and transcriptional CDKs 7 and 9, ultimately resulting in arrest at G1/S and G2/M, downregulation of the transcriptional apparatus, and repression of anti-apoptotic proteins. Considerably enhanced tumor cell apoptosis was achieved following combined treatment with 10 nmol/L roniciclib and 2.0 μmol/L cisplatin; this combination therapy achieved a response over 250% greater than either drug alone. Although roniciclib chemosensitized NPC cells to cisplatin, it did not sensitize untransformed (NP69) cells. The administration of 0.5 mg/kg roniciclib to BALB/c xenograft mice was well tolerated and effectively restrained tumor growth comparable to treatment with 6 mg/kg cisplatin, whereas combining these two agents produced far greater tumor suppression than either of the monotherapies. In summary, these data demonstrate that roniciclib has strong anti-NPC activity and synergizes with cisplatin chemotherapy at clinically relevant doses, thus justifying further evaluation of this combinatorial approach in clinical settings. Nasopharyngeal carcinoma (NPC) is an uncommon malignancy arising from the nasopharynx epithelium, and is endemic to east and southeast parts of Asia where they account for 70% of worldwide incidence. Researchers from the Cancer Science Institute of Singapore examined the anti-tumor effects of roniciclib—a small-molecule drug that blocks a family of enzymes known as cyclin-dependent kinases (CDKs) which are classically involved in cell cycle progression and transcription—in cell lines and mouse models of nasopharyngeal carcinoma. Because CDK/cyclin complexes have a putative role in DNA repair, roniciclib was combined with cisplatin, a DNA-damaging agent which is currently used in chemotherapy of NPC. The authors found that roniciclib had potent anti-NPC effects when given alone, whereas the combination of roniciclib and cisplatin proved to be highly synergistic and restrained tumor growth to a greater extent than either drugs given alone. Interestingly, roniciclib appeared to selectively enhance the anti-cancer effects of cisplatin in cancerous cells while this “chemo-sensitizing” phenomenon was not seen in non-cancerous cells, suggesting that giving both drugs together could improve the effectiveness of standard chemotherapy without incurring additional toxicities. These findings suggest that roniciclib should be evaluated clinically in patients with NPC.
Collapse
Affiliation(s)
- Nicholas L Syn
- 1Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,2Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Pei Li Lim
- 1Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Li Ren Kong
- 1Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Lingzhi Wang
- 1Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,3Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Andrea Li-Ann Wong
- 1Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,2Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Chwee Ming Lim
- 4Department of Otolaryngology-Head and Neck Surgery, National University Health System, Singapore, Singapore
| | - Thomas Kwok Seng Loh
- 4Department of Otolaryngology-Head and Neck Surgery, National University Health System, Singapore, Singapore
| | | | - Boon Cher Goh
- 1Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,2Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,3Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Wen-Son Hsieh
- 1Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| |
Collapse
|
31
|
Pan-HDAC inhibition by panobinostat mediates chemosensitization to carboplatin in non-small cell lung cancer via attenuation of EGFR signaling. Cancer Lett 2018; 417:152-160. [DOI: 10.1016/j.canlet.2017.12.030] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 12/02/2017] [Accepted: 12/15/2017] [Indexed: 01/04/2023]
|
32
|
Kumarakulasinghe NB, Syn N, Soon YY, Asmat A, Zheng H, Loy EY, Pang B, Soo RA. EGFR kinase inhibitors and gastric acid suppressants in EGFR-mutant NSCLC: a retrospective database analysis of potential drug interaction. Oncotarget 2018; 7:85542-85550. [PMID: 27907909 PMCID: PMC5356756 DOI: 10.18632/oncotarget.13458] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 10/27/2016] [Indexed: 11/25/2022] Open
Abstract
Background Erlotinib and gefitinib are weak base drugs whose absorption and clinical efficacy may be impaired by concomitant gastric acid suppressive (AS) therapy, yet proton pump inhibitors (PPIs) and histamine-2 receptor antagonists (H2As) are widely indicated in non-small cell lung cancer (NSCLC) patients for the prevention and treatment of erlotinib-induced gastrointestinal injury and corticosteroid-associated gastric irritation. We assessed the clinical relevance of this potential drug-drug interaction (DDI) in a retrospective cohort of EGFR-mutant NSCLC patients. Results The AS usage rate was 35%. In the overall cohort, AS users did not experience poorer OS (HR: 1.47, 95% CI: 0.92 – 2.35, P = 0.10; median, 11.4 versus 17.5 months) or PFS (HR = 1.37, 95% CI: 0.89 – 2.12, P = 0.16; median, 7.6 versus 8.7 months) compared with non-users in multivariate Cox regression analysis. However, subgroup analyses indicated that AS usage was associated with significantly poorer OS and PFS in patients who had fewer or milder comorbidities (Charlson comorbidity index ≤ 2), those with Karnofsky performance status < 90, and never-smokers. Materials and Methods A retrospective database analysis of 157 patients given erlotinib or gefitinib for EGFR-mutant advanced NSCLC from two institutions was conducted. Patients were classified as AS-users if the periods of AS and anti-EGFR therapy overlapped by ≥ 30%. Overall survival (OS) and progression-free survival (PFS) were assessed according to AS usage. Conclusions Concomitant AS therapy did not have an adverse impact on OS and/or PFS in the overall cohort. Our subgroup findings should be regarded exploratory and require replication in a large prospective cohort.
Collapse
Affiliation(s)
| | - Nicholas Syn
- Department of Haematology-Oncology, National University Cancer Institute, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yu Yang Soon
- Department of Radiation Oncology, National University Cancer Institute, Singapore
| | - Atasha Asmat
- Department of General Surgery, Tan Tock Seng Hospital, Singapore
| | - Huili Zheng
- National Registry of Diseases Office, Health Promotion Board, Singapore
| | - En Yun Loy
- National Registry of Diseases Office, Health Promotion Board, Singapore
| | - Brendan Pang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Pathology, National University Health System, Singapore
| | - Ross Andrew Soo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| |
Collapse
|
33
|
Macías M, Alegre E, Díaz-Lagares A, Patiño A, Pérez-Gracia JL, Sanmamed M, López-López R, Varo N, González A. Liquid Biopsy: From Basic Research to Clinical Practice. Adv Clin Chem 2017; 83:73-119. [PMID: 29304904 DOI: 10.1016/bs.acc.2017.10.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Liquid biopsy refers to the molecular analysis in biological fluids of nucleic acids, subcellular structures, especially exosomes, and, in the context of cancer, circulating tumor cells. In the last 10 years, there has been an intensive research in liquid biopsy to achieve a less invasive and more precise personalized medicine. Molecular assessment of these circulating biomarkers can complement or even surrogate tissue biopsy. Because of this research, liquid biopsy has been introduced in clinical practice, especially in oncology, prenatal screening, and transplantation. Here we review the biology, methodological approaches, and clinical applications of the main biomarkers involved in liquid biopsy.
Collapse
Affiliation(s)
| | - Estibaliz Alegre
- Clínica Universidad de Navarra, Pamplona, Spain; The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Angel Díaz-Lagares
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS), CIBERONC, Santiago de Compostela, Spain; Roche-CHUS Joint Unit, University Clinical Hospital of Santiago (CHUS), Santiago de Compostela, Spain
| | - Ana Patiño
- Clínica Universidad de Navarra, Pamplona, Spain; The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Jose L Pérez-Gracia
- Clínica Universidad de Navarra, Pamplona, Spain; The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Miguel Sanmamed
- Yale University School of Medicine, New Haven, CT, United States
| | - Rafael López-López
- Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS), CIBERONC, Santiago de Compostela, Spain; Roche-CHUS Joint Unit, University Clinical Hospital of Santiago (CHUS), Santiago de Compostela, Spain
| | - Nerea Varo
- Clínica Universidad de Navarra, Pamplona, Spain; The Health Research Institute of Navarra (IDISNA), Pamplona, Spain
| | - Alvaro González
- Clínica Universidad de Navarra, Pamplona, Spain; The Health Research Institute of Navarra (IDISNA), Pamplona, Spain.
| |
Collapse
|
34
|
Mehrotra M, Singh RR, Loghavi S, Duose DY, Barkoh BA, Behrens C, Patel KP, Routbort MJ, Kopetz S, Broaddus RR, Medeiros LJ, Wistuba II, Luthra R. Detection of somatic mutations in cell-free DNA in plasma and correlation with overall survival in patients with solid tumors. Oncotarget 2017. [PMID: 29535804 PMCID: PMC5828199 DOI: 10.18632/oncotarget.21982] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
A suitable clinical-grade platform is required for detection of somatic mutations with high sensitivity in cell-free DNA (cfDNA) of patients with solid tumors. In this study, we evaluated in parallel ultra-deep NGS with MassARRAY and allele-specific droplet digital PCR (ddPCR) for cfDNA genotyping and correlated cfDNA yield and mutation status with overall survival (OS) of patients. We assessed plasma samples from 46 patients with various advanced metastatic solid tumors and known mutations by deep sequencing using an Ampliseq cancer hotspot panel V2 on Ion Proton. A subset of these samples with DNA availability was tested by ddPCR and UltraSEEK MassARRAY for mutation detection in 5 genes (IDH1, PIK3CA, KRAS, BRAF, and NRAS). Sixty one of 104 expected tissue mutations and 6 additional mutations not present in the tissue were detected in cfDNA. ddPCR and MassARRAY showed 83% and 77% concordance with NGS for mutation detection with 100% and 79% sensitivity, respectively. The median OS of patients with lower cfDNA yield (74 vs 50 months; P < 0.03) and cfDNA negative for mutations (74.2 vs 53 months; p < 0.04) was significantly longer than in patients with higher cfDNA yield and positive for mutations. A limit-of-detection of 0.1% was demonstrated for ddPCR and MassARRAY platforms using a serially diluted positive cfDNA sample. The MassARRAY and ddPCR systems enable fast and cost-effective genotyping for a targeted set of mutations and can be used for single gene testing to guide response to chemotherapy or for orthogonal validation of NGS results.
Collapse
Affiliation(s)
- Meenakshi Mehrotra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rajesh R Singh
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sanam Loghavi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dzifa Yawa Duose
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bedia A Barkoh
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Carmen Behrens
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Keyur P Patel
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mark J Routbort
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Russell R Broaddus
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rajyalakshmi Luthra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
35
|
Heong V, Syn NL, Lee XW, Sapari NS, Koh XQ, Adam Isa ZF, Sy Lim J, Lim D, Pang B, Thian YL, Ng LK, Wong AL, Soo RA, Yong WP, Chee CE, Lee SC, Goh BC, Soong R, Tan DSP. Value of a molecular screening program to support clinical trial enrollment in Asian cancer patients: The Integrated Molecular Analysis of Cancer (IMAC) Study. Int J Cancer 2017; 142:1890-1900. [PMID: 28994108 DOI: 10.1002/ijc.31091] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/15/2017] [Accepted: 09/27/2017] [Indexed: 02/01/2023]
Abstract
The value of precision oncology initiatives in Asian contexts remains unresolved. Here, we review the institutional implementation of prospective molecular screening to facilitate accrual of patients into biomarker-driven clinical trials, and to explore the mutational landscape of advanced tumors occurring in a prospective cohort of Asian patients (n = 396) with diverse cancer types. Next-generation sequencing (NGS) and routine clinicopathological assays, such as immunohistochemistry, copy number analysis and in situ hybridization tests, were performed on tumor samples. Actionable biomarker results were used to identify eligibility for early-phase, biomarker-driven clinical trials. Overall, NGS was successful in 365 of 396 patients (92%), achieving a mean depth of 1,943× and coverage uniformity of 96%. The median turnaround time from sample receipt to return of genomic results was 26.0 days (IQR, 19.0-39.0 days). Reportable mutations were found in 300 of 365 patients (82%). Ninety-one percent of patients at study enrollment indicated consent to receive incidental findings and willingness to undergo genetic counseling if required. The most commonly mutated oncogenes included KRAS (19%), PIK3CA (16%), EGFR (5%), BRAF (3%) and KIT (3%); while the most frequently mutated tumor suppressor genes included TP53 (40%), SMARCB1 (12%), APC (8%), PTEN (6%) and SMAD4 (5%). Among 23 patients enrolled in genotype-matched trials, median progression-free survival was 2.9 months (IQR, 1.5-4.0 months). Nine of 20 evaluable patients (45%; 95% CI, 23.1-68.5%) derived clinical benefit, including 3 partial responses and 6 with stable disease lasting ≥ 8 weeks.
Collapse
Affiliation(s)
- Valerie Heong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Nicholas L Syn
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Xiao Wen Lee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Nur Sabrina Sapari
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Xue Qing Koh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Zul Fazreen Adam Isa
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Joey Sy Lim
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Diana Lim
- Department of Pathology, Yong Loo Lin School of Medicine, National University Health System, Singapore.,Department of Pathology, National University Hospital, National University Health System, Singapore
| | - Brendan Pang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Pathology, Yong Loo Lin School of Medicine, National University Health System, Singapore.,Department of Pathology, National University Hospital, National University Health System, Singapore
| | - Yee Liang Thian
- Department of Diagnostic Imaging, National University Hospital, National University Health System, Singapore
| | - Lai Kuan Ng
- Department of Pathology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Andrea L Wong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Ross Andrew Soo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Wei Peng Yong
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Cheng Ean Chee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Soo-Chin Lee
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Boon-Cher Goh
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Richie Soong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Pathology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - David S P Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University Health System, Singapore
| |
Collapse
|
36
|
Roudi R, Syn NL, Roudbary M. Antimicrobial Peptides As Biologic and Immunotherapeutic Agents against Cancer: A Comprehensive Overview. Front Immunol 2017; 8:1320. [PMID: 29081781 PMCID: PMC5645638 DOI: 10.3389/fimmu.2017.01320] [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: 08/26/2017] [Accepted: 09/29/2017] [Indexed: 12/15/2022] Open
Abstract
Antimicrobial peptides (AMPs) are a pervasive and evolutionarily ancient component of innate host defense which is present in virtually all classes of life. In recent years, evidence has accumulated that parallel or de novo mechanisms by which AMPs curb infectious pathologies are also effective at restraining cancer cell proliferation and dissemination, and have consequently stimulated significant interest in their deployment as novel biologic and immunotherapeutic agents against human malignancies. In this review, we explicate the biochemical underpinnings of their tumor-selectivity, and discuss results of recent clinical trials (outside of oncologic indications) which substantiate their safety and tolerability profiles. Next, we present evidence for their preclinical antitumor activity, systematically organized by the major and minor classes of natural AMPs. Finally, we discuss the barriers to their clinical implementation and envision directions for further development.
Collapse
Affiliation(s)
- Raheleh Roudi
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Nicholas L Syn
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Maryam Roudbary
- Department of Medical Mycology and Parasitology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Ma R, Xu H, Wu J, Sharma A, Bai S, Dun B, Jing C, Cao H, Wang Z, She JX, Feng J. Identification of serum proteins and multivariate models for diagnosis and therapeutic monitoring of lung cancer. Oncotarget 2017; 8:18901-18913. [PMID: 28121629 PMCID: PMC5386656 DOI: 10.18632/oncotarget.14782] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 12/27/2016] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is one of the most prevalent cancers and has very poor treatment outcome. Biomarkers useful for screening and assessing early therapeutic response may significantly improve the therapeutic outcome but are still lacking. In this study, serum samples from 218 non-small cell lung cancer (NSCLC) patients, 34 small cell lung cancer (SCLC) patients and 171 matched healthy controls from China were analyzed for 11 proteins using the Luminex multiplex assay. Eight of the 11 proteins (OPN, SAA, CRP, CYFRA21.1, CEA, NSE, AGP and HGF) are significantly elevated in NSCLC and SCLC (p = 10−5−10−59). At the individual protein level, OPN has the best diagnostic value for NSCLC (AUC = 0.92), two acute phase proteins (SAA and CRP) have AUC near 0.83, while CEA and CYFRA21.1 also possess good AUC (0.81 and 0.77, respectively). More importantly, several three-protein combinations that contain OPN and CEA plus one of four proteins (CRP, SAA, CYFRA21.1 or NSE) have excellent diagnostic potential for NSCLC (AUC = 0.96). Four proteins (CYFRA21.1, CRP, SAA and NSE) are severely reduced and three proteins (OPN, MIF and NSE) are moderately decreased after platinum-based chemotherapy. Therapeutic response index (TRI) computed with 3–5 proteins suggests that approximately 25% of the NSCLC patients respond well to the therapy and TRI is significantly correlated with pre-treatment protein levels. Our data suggest that therapeutic response in NSCLC patients can be effectively measured but personalized biomarkers may be needed to monitor different subsets of patients.
Collapse
Affiliation(s)
- Rong Ma
- Clinical Cancer Research Center, Jiangsu Cancer Hospital, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Nanjing, Jiangsu, 210009, China
| | - Heng Xu
- Jiangsu Province Institute of Materia Medica, Nanjing Tech University, Nanjing 211816, China
| | - Jianzhong Wu
- Clinical Cancer Research Center, Jiangsu Cancer Hospital, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Nanjing, Jiangsu, 210009, China
| | - Ashok Sharma
- Center for Biotechnology and Genomic Medicine, and Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Shan Bai
- Center for Biotechnology and Genomic Medicine, and Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Boying Dun
- Center for Biotechnology and Genomic Medicine, and Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Changwen Jing
- Clinical Cancer Research Center, Jiangsu Cancer Hospital, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Nanjing, Jiangsu, 210009, China
| | - Haixia Cao
- Clinical Cancer Research Center, Jiangsu Cancer Hospital, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Nanjing, Jiangsu, 210009, China
| | - Zhuo Wang
- Clinical Cancer Research Center, Jiangsu Cancer Hospital, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Nanjing, Jiangsu, 210009, China
| | - Jin-Xiong She
- Center for Biotechnology and Genomic Medicine, and Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Jifeng Feng
- Clinical Cancer Research Center, Jiangsu Cancer Hospital, Nanjing Medical University Affiliated Cancer Hospital Cancer Institute of Jiangsu Province, Nanjing, Jiangsu, 210009, China
| |
Collapse
|
38
|
Wagner AD, Syn NLX, Moehler M, Grothe W, Yong WP, Tai B, Ho J, Unverzagt S. Chemotherapy for advanced gastric cancer. Cochrane Database Syst Rev 2017; 8:CD004064. [PMID: 28850174 PMCID: PMC6483552 DOI: 10.1002/14651858.cd004064.pub4] [Citation(s) in RCA: 366] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Gastric cancer is the fifth most common cancer worldwide. In "Western" countries, most people are either diagnosed at an advanced stage, or develop a relapse after surgery with curative intent. In people with advanced disease, significant benefits from targeted therapies are currently limited to HER-2 positive disease treated with trastuzumab, in combination with chemotherapy, in first-line. In second-line, ramucirumab, alone or in combination with paclitaxel, demonstrated significant survival benefits. Thus, systemic chemotherapy remains the mainstay of treatment for advanced gastric cancer. Uncertainty remains regarding the choice of the regimen. OBJECTIVES To assess the efficacy of chemotherapy versus best supportive care (BSC), combination versus single-agent chemotherapy and different chemotherapy combinations in advanced gastric cancer. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials, MEDLINE and Embase up to June 2016, reference lists of studies, and contacted pharmaceutical companies and experts to identify randomised controlled trials (RCTs). SELECTION CRITERIA We considered only RCTs on systemic, intravenous or oral chemotherapy versus BSC, combination versus single-agent chemotherapy and different chemotherapy regimens in advanced gastric cancer. DATA COLLECTION AND ANALYSIS Two review authors independently identified studies and extracted data. A third investigator was consulted in case of disagreements. We contacted study authors to obtain missing information. MAIN RESULTS We included 64 RCTs, of which 60 RCTs (11,698 participants) provided data for the meta-analysis of overall survival. We found chemotherapy extends overall survival (OS) by approximately 6.7 months more than BSC (hazard ratio (HR) 0.3, 95% confidence intervals (CI) 0.24 to 0.55, 184 participants, three studies, moderate-quality evidence). Combination chemotherapy extends OS slightly (by an additional month) versus single-agent chemotherapy (HR 0.84, 95% CI 0.79 to 0.89, 4447 participants, 23 studies, moderate-quality evidence), which is partly counterbalanced by increased toxicity. The benefit of epirubicin in three-drug combinations, in which cisplatin is replaced by oxaliplatin and 5-FU is replaced by capecitabine is unknown.Irinotecan extends OS slightly (by an additional 1.6 months) versus non-irinotecan-containing regimens (HR 0.87, 95% CI 0.80 to 0.95, 2135 participants, 10 studies, high-quality evidence).Docetaxel extends OS slightly (just over one month) compared to non-docetaxel-containing regimens (HR 0.86, 95% CI 0.78 to 0.95, 2001 participants, eight studies, high-quality evidence). However, due to subgroup analyses, we are uncertain whether docetaxel-containing combinations (docetaxel added to a single-agent or two-drug combination) extends OS due to moderate-quality evidence (HR 0.80, 95% CI 0.71 to 0.91, 1466 participants, four studies, moderate-quality evidence). When another chemotherapy was replaced by docetaxel, there is probably little or no difference in OS (HR 1.05; 0.87 to 1.27, 479 participants, three studies, moderate-quality evidence). We found there is probably little or no difference in OS when comparing capecitabine versus 5-FU-containing regimens (HR 0.94, 95% CI 0.79 to 1.11, 732 participants, five studies, moderate-quality evidence) .Oxaliplatin may extend (by less than one month) OS versus cisplatin-containing regimens (HR 0.81, 95% CI 0.67 to 0.98, 1105 participants, five studies, low-quality evidence). We are uncertain whether taxane-platinum combinations with (versus without) fluoropyrimidines extend OS due to very low-quality evidence (HR 0.86, 95% CI 0.71 to 1.06, 482 participants, three studies, very low-quality evidence). S-1 regimens improve OS slightly (by less than an additional month) versus 5-FU-containing regimens (HR 0.91, 95% CI 0.83 to 1.00, 1793 participants, four studies, high-quality evidence), however since S-1 is used in different doses and schedules between Asian and non-Asian population, the applicability of this finding to individual populations is uncertain. AUTHORS' CONCLUSIONS Chemotherapy improves survival (by an additional 6.7 months) in comparison to BSC, and combination chemotherapy improves survival (by an additional month) compared to single-agent 5-FU. Testing all patients for HER-2 status may help to identify patients with HER-2-positive tumours, for whom, in the absence of contraindications, trastuzumab in combination with capecitabine or 5-FU in combination with cisplatin has been shown to be beneficial. For HER-2 negative people, all different two-and three-drug combinations including irinotecan, docetaxel, oxaliplatin or oral 5-FU prodrugs are valid treatment options for advanced gastric cancer, and consideration of the side effects of each regimen is essential in the treatment decision. Irinotecan-containing combinations and docetaxel-containing combinations (in which docetaxel was added to a single-agent or two-drug (platinum/5-FUcombination) show significant survival benefits in the comparisons studied above. Furthermore, docetaxel-containing three-drug regimens have increased response rates, but the advantages of the docetaxel-containing three-drug combinations (DCF, FLO-T) are counterbalanced by increased toxicity. Additionally, oxaliplatin-containing regimens demonstrated a benefit in OS as compared to the same regimen containing cisplatin, and there is a modest survival improvement of S-1 compared to 5-FU-containing regimens.Whether the survival benefit for three-drug combinations including cisplatin, 5-FU, and epirubicin as compared to the same regimen without epirubicin is still valid when second-line therapy is routinely administered and when cisplatin is replaced by oxaliplatin and 5-FU by capecitabine is questionable. Furthermore, the magnitude of the observed survival benefits for the three-drug regimens is not large enough to be clinically meaningful as defined recently by the American Society for Clinical Oncology (Ellis 2014). In contrast to the comparisons in which a survival benefit was observed by adding a third drug to a two-drug regimen at the cost of increased toxicity, the comparison of regimens in which another chemotherapy was replaced by irinotecan was associated with a survival benefit (of borderline statistical significance), but without increased toxicity. For this reason irinotecan/5-FU-containing combinations are an attractive option for first-line treatment. Although they need to be interpreted with caution, subgroup analyses of one study suggest that elderly people have a greater benefit form oxaliplatin, as compared to cisplatin-based regimens, and that people with locally advanced disease or younger than 65 years might benefit more from a three-drug regimen including 5-FU, docetaxel, and oxaliplatin as compared to a two-drug combination of 5-FU and oxaliplatin, a hypothesis that needs further confirmation. For people with good performance status, the benefit of second-line chemotherapy has been established in several RCTs.
Collapse
Affiliation(s)
- Anna Dorothea Wagner
- Lausanne University Hospitals and ClinicsDepartment of OncologyRue du Bugnon 46LausanneSwitzerland1011
| | - Nicholas LX Syn
- National University Cancer InstituteDepartment of Haematology‐Oncology1E Kent Ridge RoadNUHS Tower Block, Level 7SingaporeSingapore119228
| | - Markus Moehler
- University Medical Center of the Johannes Gutenberg UniversityDepartment of Internal MedicineLangenbeckstrasse 1MainzGermany55131
| | - Wilfried Grothe
- Martin‐Luther‐University Halle‐WittenbergDepartment of Internal Medicine IErnst‐Grube‐Str. 40Halle/SaaleGermany06097
| | - Wei Peng Yong
- National University Cancer InstituteDepartment of Haematology‐Oncology1E Kent Ridge RoadNUHS Tower Block, Level 7SingaporeSingapore119228
| | - Bee‐Choo Tai
- National University of SingaporeSaw Swee Hock School of Public Health12 Science Drive 2#10‐03FSingaporeSingapore117549
| | - Jingshan Ho
- National University Cancer InstituteDepartment of Haematology‐Oncology1E Kent Ridge RoadNUHS Tower Block, Level 7SingaporeSingapore119228
| | - Susanne Unverzagt
- Martin‐Luther‐University Halle‐WittenbergInstitute of Medical Epidemiology, Biostatistics and InformaticsMagdeburge Straße 8Halle/SaaleGermany06097
| | | |
Collapse
|
39
|
Siniard RC, Harada S. Immunogenomics: using genomics to personalize cancer immunotherapy. Virchows Arch 2017; 471:209-219. [DOI: 10.1007/s00428-017-2140-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/21/2017] [Accepted: 04/27/2017] [Indexed: 01/06/2023]
|
40
|
Syn NL, Wang L, Chow EKH, Lim CT, Goh BC. Exosomes in Cancer Nanomedicine and Immunotherapy: Prospects and Challenges. Trends Biotechnol 2017; 35:665-676. [PMID: 28365132 DOI: 10.1016/j.tibtech.2017.03.004] [Citation(s) in RCA: 295] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 02/27/2017] [Accepted: 03/02/2017] [Indexed: 12/13/2022]
Abstract
Exosomes (versatile, cell-derived nanovesicles naturally endowed with exquisite target-homing specificity and the ability to surmount in vivo biological barriers) hold substantial promise for developing exciting approaches in drug delivery and cancer immunotherapy. Specifically, bioengineered exosomes are being successfully deployed to deliver potent tumoricidal drugs (siRNAs and chemotherapeutic compounds) preferentially to cancer cells, while a new generation of exosome-based therapeutic cancer vaccines has produced enticing results in early-phase clinical trials. Here, we review the state-of-the-art technologies and protocols, and discuss the prospects and challenges for the clinical development of this emerging class of therapeutics.
Collapse
Affiliation(s)
- Nicholas L Syn
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Hematology-Oncology, National University Cancer Institute, Singapore
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore.
| | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Chwee Teck Lim
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology (SMART) Centre, Singapore; Mechanobiology Institute, National University of Singapore, Singapore; Department of Biomedical Engineering, National University of Singapore, Singapore; Department of Mechanical Engineering, National University of Singapore, Singapore
| | - Boon-Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Hematology-Oncology, National University Cancer Institute, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore; Developmental Therapeutics Unit, National University Cancer Institute, Singapore
| |
Collapse
|
41
|
Functional Roles and Therapeutic Applications of Exosomes in Hepatocellular Carcinoma. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2931813. [PMID: 28265569 PMCID: PMC5318635 DOI: 10.1155/2017/2931813] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 12/15/2016] [Accepted: 01/04/2017] [Indexed: 12/21/2022]
Abstract
Exosomes are important in intercellular communication. They assure the horizontal transfer of specific functional contents (i.e., proteins, lipids, RNA molecules, and circulating DNA) from donor to recipient cells. Notably, tumor-derived exosomes (TDEs) appear to be an important vehicle of specific signals in cancer, impacting on tumor growth and metastasis. Recent researches point to the characterization of exosomes in Hepatocellular Carcinoma (HCC), the major adult liver malignancy. In this review, we summarize current findings on HCC exosomes, focusing on the identification of noncoding RNAs as exosome-enriched functional regulators and new potential biomarkers. The great potential of exosomes in future HCC diagnostic and therapeutic approaches is underlined.
Collapse
|
42
|
Bonifati V. Will New Genetic Techniques Like Exome Sequencing and Others Obviate the Need for Clinical Expertise? Yes. Mov Disord Clin Pract 2017; 4:36-38. [PMID: 30713946 DOI: 10.1002/mdc3.12438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/08/2016] [Accepted: 08/12/2016] [Indexed: 12/24/2022] Open
Affiliation(s)
- Vincenzo Bonifati
- Department of Clinical Genetics Erasmus Medical Center Rotterdam the Netherlands
| |
Collapse
|
43
|
Yildirim O, Gottwald M, Schüler P, Michel MC. Opportunities and Challenges for Drug Development: Public-Private Partnerships, Adaptive Designs and Big Data. Front Pharmacol 2016; 7:461. [PMID: 27999543 PMCID: PMC5138214 DOI: 10.3389/fphar.2016.00461] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 11/16/2016] [Indexed: 01/18/2023] Open
Abstract
Drug development faces the double challenge of increasing costs and increasing pressure on pricing. To avoid that lack of perceived commercial perspective will leave existing medical needs unmet, pharmaceutical companies and many other stakeholders are discussing ways to improve the efficiency of drug Research and Development. Based on an international symposium organized by the Medical School of the University of Duisburg-Essen (Germany) and held in January 2016, we discuss the opportunities and challenges of three specific areas, i.e., public–private partnerships, adaptive designs and big data. Public–private partnerships come in many different forms with regard to scope, duration and type and number of participants. They range from project-specific collaborations to strategic alliances to large multi-party consortia. Each of them offers specific opportunities and faces distinct challenges. Among types of collaboration, investigator-initiated studies are becoming increasingly popular but have legal, ethical, and financial implications. Adaptive trial designs are also increasingly discussed. However, adaptive should not be used as euphemism for the repurposing of a failed trial; rather it requires carefully planning and specification before a trial starts. Adaptive licensing can be a counter-part of adaptive trial design. The use of Big Data is another opportunity to leverage existing information into knowledge useable for drug discovery and development. Respecting limitations of informed consent and privacy is a key challenge in the use of Big Data. Speakers and participants at the symposium were convinced that appropriate use of the above new options may indeed help to increase the efficiency of future drug development.
Collapse
Affiliation(s)
- Oktay Yildirim
- Institute of Pharmacology, University Duisburg-Essen Essen, Germany
| | | | - Peter Schüler
- Department of Drug Development Services CNS, ICON Clinical Research Langen, Germany
| | - Martin C Michel
- Department of Pharmacology, Johannes Gutenberg University Mainz, Germany
| |
Collapse
|
44
|
Bertier G, Carrot-Zhang J, Ragoussis V, Joly Y. Integrating precision cancer medicine into healthcare-policy, practice, and research challenges. Genome Med 2016; 8:108. [PMID: 27776531 PMCID: PMC5075982 DOI: 10.1186/s13073-016-0362-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Precision medicine (PM) can be defined as a predictive, preventive, personalized, and participatory healthcare service delivery model. Recent developments in molecular biology and information technology make PM a reality today through the use of massive amounts of genetic, ‘omics’, clinical, environmental, and lifestyle data. With cancer being one of the most prominent public health threats in developed countries, both the research community and governments have been investing significant time, money, and efforts in precision cancer medicine (PCM). Although PCM research is extremely promising, a number of hurdles still remain on the road to an optimal integration of standardized and evidence-based use of PCM in healthcare systems. Indeed, PCM raises a number of technical, organizational, ethical, legal, social, and economic challenges that have to be taken into account in the development of an appropriate health policy framework. Here, we highlight some of the more salient issues regarding the standards needed for integration of PCM into healthcare systems, and we identify fields where more research is needed before policy can be implemented. Key challenges include, but are not limited to, the creation of new standards for the collection, analysis, and sharing of samples and data from cancer patients, and the creation of new clinical trial designs with renewed endpoints. We believe that these issues need to be addressed as a matter of priority by public health policymakers in the coming years for a better integration of PCM into healthcare.
Collapse
Affiliation(s)
- Gabrielle Bertier
- Center of Genomics and Policy, McGill University, 740 Dr. Penfield Avenue, Montreal, QC, H3A 0G1, Canada. .,Université Toulouse III Paul Sabatier and Inserm UMR 102, 37 allées Jules Guesde, F-31000, Toulouse, France.
| | - Jian Carrot-Zhang
- Center of Genomics and Policy, McGill University, 740 Dr. Penfield Avenue, Montreal, QC, H3A 0G1, Canada
| | - Vassilis Ragoussis
- Sargent College, Boston University, One Silber Way, Boston, MA, 02215, USA
| | - Yann Joly
- Center of Genomics and Policy, McGill University, 740 Dr. Penfield Avenue, Montreal, QC, H3A 0G1, Canada
| |
Collapse
|