1
|
Haffner MR, Saiz AM, Darrow MA, Judge SJ, Laun T, Arora A, Taylor SL, Randall RL, Alvarez EM, Thorpe SW. Effect of ALDH1A1 and CD44 on Survival and Disease Recurrence in Patients With Osteosarcoma. Cureus 2024; 16:e52404. [PMID: 38371078 PMCID: PMC10869251 DOI: 10.7759/cureus.52404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2024] [Indexed: 02/20/2024] Open
Abstract
PURPOSE Emerging evidence suggests that osteosarcoma stem cells (OSCs) may be responsible for tumor initiation propagation, recurrence, and resistance to therapy. We set out to evaluate the relationship between the abundance of ALDH1A1 and CD44-positive cells in biopsy and resection samples on disease recurrence and overall survival. METHODS A retrospective review of 20 patients, including biopsy and resection samples, was performed at a comprehensive cancer center. Additionally, we queried the publicly available TARGET dataset of osteosarcoma patients. RESULTS Neither the percentages of ALDH1A1-positive cells nor CD44-positive cells were significantly associated with overall mortality or disease recurrence in either biopsy or resection samples. Unlike our institutional data, overall survival was significantly correlated to higher ALDH1A1 expression in the TARGET dataset both in univariate and age-adjusted analyses. CONCLUSIONS ADLH1 and CD44, potential markers of OSCs, were not found to be reliable clinical immunohistochemical prognostic markers for osteosarcoma patient survival, specifically disease-free survival. Osteosarcoma patients with high ALDH1A1 RNA expression showed improved overall survival in examining a national genomic database of osteosarcoma patients but again no association with disease-free survival. The potential of CD44 and ALDH1A1 as cellular-specific prognostic markers of survival, and as possible molecular targets, may be limited in osteosarcoma.
Collapse
Affiliation(s)
- Max R Haffner
- Orthopedic Surgery, UC (University of California) Davis Health, Sacramento, USA
| | - Augustine M Saiz
- Orthopedic Surgery, UC (University of California) Davis Health, Sacramento, USA
| | - Morgan A Darrow
- Pathology and Laboratory Medicine, UC (University of California) Davis Health, Sacramento, USA
| | - Sean J Judge
- Surgery, UC (University of California) Davis Health, Sacramento, USA
| | - Tammy Laun
- Medicine, Oakland University William Beaumont School of Medicine, Auburn Hills, USA
| | - Aman Arora
- Urology, UC (University of California) Davis School of Medicine, Sacramento, USA
| | - Sandra L Taylor
- Division of Biostatistics, Public Health Sciences, Sacramento, USA
| | - R Lor Randall
- Orthopedic Surgery, UC (University of California) Davis Health, Sacramento, USA
| | - Elysia M Alvarez
- Pediatric Hematology and Oncology, UC (University of California) Davis Health, Sacramento, USA
| | - Steven W Thorpe
- Orthopedic Surgery, UC (University of California) Davis Health, Sacramento, USA
| |
Collapse
|
2
|
Al-Shamma SA, Zaher DM, Hersi F, Abu Jayab NN, Omar HA. Targeting aldehyde dehydrogenase enzymes in combination with chemotherapy and immunotherapy: An approach to tackle resistance in cancer cells. Life Sci 2023; 320:121541. [PMID: 36870386 DOI: 10.1016/j.lfs.2023.121541] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/19/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023]
Abstract
Modern cancer chemotherapy originated in the 1940s, and since then, many chemotherapeutic agents have been developed. However, most of these agents show limited response in patients due to innate and acquired resistance to therapy, which leads to the development of multi-drug resistance to different treatment modalities, leading to cancer recurrence and, eventually, patient death. One of the crucial players in inducing chemotherapy resistance is the aldehyde dehydrogenase (ALDH) enzyme. ALDH is overexpressed in chemotherapy-resistant cancer cells, which detoxifies the generated toxic aldehydes from chemotherapy, preventing the formation of reactive oxygen species and, thus, inhibiting the induction of oxidative stress and the stimulation of DNA damage and cell death. This review discusses the mechanisms of chemotherapy resistance in cancer cells promoted by ALDH. In addition, we provide detailed insight into the role of ALDH in cancer stemness, metastasis, metabolism, and cell death. Several studies investigated targeting ALDH in combination with other treatments as a potential therapeutic regimen to overcome resistance. We also highlight novel approaches in ALDH inhibition, including the potential synergistic employment of ALDH inhibitors in combination with chemotherapy or immunotherapy against different cancers, including head and neck, colorectal, breast, lung, and liver.
Collapse
Affiliation(s)
- Salma A Al-Shamma
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Dana M Zaher
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Fatema Hersi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Nour N Abu Jayab
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hany A Omar
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| |
Collapse
|
3
|
Dhanyamraju PK, Schell TD, Amin S, Robertson GP. Drug-Tolerant Persister Cells in Cancer Therapy Resistance. Cancer Res 2022; 82:2503-2514. [PMID: 35584245 PMCID: PMC9296591 DOI: 10.1158/0008-5472.can-21-3844] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/15/2022] [Accepted: 05/09/2022] [Indexed: 01/21/2023]
Abstract
One of the current stumbling blocks in our fight against cancer is the development of acquired resistance to therapy, which is attributable to approximately 90% of cancer-related deaths. Undercutting this process during treatment could significantly improve cancer management. In many cases, drug resistance is mediated by a drug-tolerant persister (DTP) cell subpopulation present in tumors, often referred to as persister cells. This review provides a summary of currently known persister cell subpopulations and approaches to target them. A specific DTP cell subpopulation with elevated levels of aldehyde dehydrogenase (ALDH) activity has stem cell-like characteristics and a high level of plasticity, enabling them to switch rapidly between high and low ALDH activity. Further studies are required to fully elucidate the functions of ALDH-high DTP cells, how they withstand drug concentrations that kill other cells, and how they rapidly adapt under levels of high cellular stress and eventually lead to more aggressive, recurrent, and drug-resistant cancer. Furthermore, this review addresses the processes used by the ALDH-high persister cell subpopulation to enable cancer progression, the ALDH isoforms important in these processes, interactions of ALDH-high DTPs with the tumor microenvironment, and approaches to therapeutically modulate this subpopulation in order to more effectively manage cancer.
Collapse
Affiliation(s)
- Pavan Kumar Dhanyamraju
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Todd D Schell
- Departments of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Shantu Amin
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Gavin P Robertson
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Pathology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Dermatology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- The Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Penn State Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| |
Collapse
|
4
|
Filipiak-Duliban A, Brodaczewska K, Majewska A, Kieda C. Spheroid culture models adequately imitate distinctive features of the renal cancer or melanoma microenvironment. In Vitro Cell Dev Biol Anim 2022; 58:349-364. [PMID: 35536385 DOI: 10.1007/s11626-022-00685-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/24/2022] [Indexed: 11/05/2022]
Abstract
Tumor development studies should adapt to cancer cells' specific mechanisms in connection with their microenvironment. Standard two-dimensional cultures and gas composition are not relevant to the real cancer environment. Existing three-dimensional models are often requiring sophisticated conditions. Here, we propose and characterize, in two cancer models, melanoma (B16F10) and kidney cancer (RenCa), a three-dimensional culture method, reporting the presence of hypoxia-related genes/proteins and aggressiveness mechanisms (epithelial mesenchymal transition and cancer stem cells). We validate the designed three-dimensional method by comparing it with in vivo growing tumors. The developed method brings simplicity and data reproducibility. Melanoma spheroid-growing cells reached a cell cycle arrest at the G0/G1 phase and showed induction of hypoxia. Spheroid-recovered RenCa cells were enriched in proliferating cells and displayed delayed hypoxia. Moreover, the responses to hypoxia observed in spheroids were validated by in vivo tumor studies for both lines. Three-dimensional shapes induced cancer stem cells in renal cancer, whereas epithelial to mesenchymal transition occurred in the melanoma model. Such distinction in the use of different aggressiveness-leading pathways was observed in in vivo melanoma vs kidney tumors. Thus, this 3D culture model approach is adequate to uncover crucial molecular pathways using distinct mechanisms to reach aggressiveness; i.e., B16F10 cells perform epithelial to mesenchymal transition while RenCa cells dedifferentiate into cancer stem cells. Such three-dimensional models help mimic the in vivo tumor features, i.e., hypoxia and aggressiveness mechanisms as validated here by next-generation sequencing analysis, and are proposed for further alternative methods to in vivo studies.
Collapse
Affiliation(s)
- Aleksandra Filipiak-Duliban
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland. .,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland.
| | - Klaudia Brodaczewska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland
| | - Aleksandra Majewska
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland.,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Claudine Kieda
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, Warsaw, Poland.,Center for Molecular Biophysics UPR 4301 CNRS, 45071, Orleans, France
| |
Collapse
|
5
|
Role of Biomarkers in the Integrated Management of Melanoma. DISEASE MARKERS 2022; 2021:6238317. [PMID: 35003391 PMCID: PMC8739586 DOI: 10.1155/2021/6238317] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/13/2021] [Indexed: 12/21/2022]
Abstract
Melanoma, which is an aggressive skin cancer, is currently the fifth and seventh most common cancer in men and women, respectively. The American Cancer Society reported that approximately 106,110 new cases of melanoma were diagnosed in the United States in 2021, with 7,180 people dying from the disease. This information could facilitate the early detection of possible metastatic lesions and the development of novel therapeutic techniques for melanoma. Additionally, early detection of malignant melanoma remains an objective of melanoma research. Recently, melanoma treatment has substantially improved, given the availability of targeted treatments and immunotherapy. These developments have highlighted the significance of identifying biomarkers for prognosis and predicting therapy response. Biomarkers included tissue protein expression, circulating DNA detection, and genetic alterations in cancer cells. Improved diagnostic and prognostic biomarkers are becoming increasingly relevant in melanoma treatment, with the development of newer and more targeted treatments. Here, the author discusses the aspects of biomarkers in the real-time management of patients with melanoma.
Collapse
|
6
|
Safai B, Wu AG, Hamby CV. Prognostic Biomarkers in Melanoma: Tailoring Treatments to the Patient. THE JOURNAL OF CLINICAL AND AESTHETIC DERMATOLOGY 2021; 14:44-48. [PMID: 35096254 PMCID: PMC8794494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
BACKGROUND It is often difficult to accurately predict how a melanoma will progress because melanomas can be so diverse in their genetic and histological makeup. OBJECTIVE We sought to characterize the current state and progression of biomedical markers towards their utilization as prognostic indicators for patients with melanoma. METHODS A literature search of the research repository databases PubMed and GoogleScholar was conducted using the following inclusion criteria: (1) published within the last 10 years, and (2) use of overall survival, disease progression, or clinical outcome as primary endpoints. Search terms included various permutations of "biomarkers," "prognostic," "immunologic," "serologic," "visual," and "melanoma." Results were evaluated for statistical power, results significance, and experimental design integrity. RESULTS The prognostic capabilities of clinical tests for malignant melanoma have made great strides in the last few years, with several serologic and immunohistochemical biomarkers being preliminarily linked to various measures of clinical prognosis. While clinical feasibility of a single sensitive and specific biomarker remains unfeasible, use of select combinations of tested biomarkers remain viable. CONCLUSION Diagnostic and prognostic genetic assays have begun to cross over from research to commercial application, giving physicians additional tools during the early stages of diagnosis to optimize and individualize treatments.
Collapse
Affiliation(s)
- Bijan Safai
- Dr. Safai is with the Department of Dermatology, Metropolitan Hospital in New York, New York
- Mr. Wu and Dr. Hamby are with New York Medical College School of Medicine in Valhalla, New York
| | - Albert G Wu
- Dr. Safai is with the Department of Dermatology, Metropolitan Hospital in New York, New York
- Mr. Wu and Dr. Hamby are with New York Medical College School of Medicine in Valhalla, New York
| | - Carl V Hamby
- Dr. Safai is with the Department of Dermatology, Metropolitan Hospital in New York, New York
- Mr. Wu and Dr. Hamby are with New York Medical College School of Medicine in Valhalla, New York
| |
Collapse
|
7
|
Liu M, Xu Y, Zhou Y, Lang R, Shi Z, Zhao J, Meng Y, Bao L. Integrated Analyses Reveal the Multi-Omics and Prognostic Characteristics of ATP5B in Breast Cancer. Front Genet 2021; 12:652474. [PMID: 34122507 PMCID: PMC8194306 DOI: 10.3389/fgene.2021.652474] [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: 01/12/2021] [Accepted: 04/19/2021] [Indexed: 11/13/2022] Open
Abstract
The beta subunit of F1Fo-ATP synthase (ATP5B) has been demonstrated to play an essential role in tumor progression and metastasis. However, there has been no comprehensive pan-cancer multi-omics analysis of ATP5B, while the clinical relevance of ATP5B and its potential mechanism in regulating breast cancer are still poorly understood. In this study, we demonstrated that ATP5B has a higher frequency of amplification than deletion in most cancer types, and the copy number variation (CNV) of ATP5B was significantly positively correlated with its mRNA expression level. DNA methylation analysis across pan-cancer also revealed a strong correlation between ATP5B expression and epigenetic changes. We identified 6 significant methylation sites involved in the regulation of ATP5B expression. Tissue microarrays (TMA) from 129 breast cancer samples, integrated with multiple additional breast cancer dataset, were used to evaluate the ATP5B expression and its correlation with prognosis. Higher levels of ATP5B expression were consistently associated with a worse OS in all datasets, and Cox regression analysis suggested that ATP5B expression was an independent prognostic factor. Gene enrichment analysis indicated that the gene signatures of DNA damage recognition, the E-cadherin nascent pathway and the PLK1 pathway were enriched in ATP5B-high patients. Moreover, somatic mutation analysis showed that a significant different mutation frequency of CDH1 and ADAMTSL3 could be observed between the ATP5B-high and ATP5B-low groups. In conclusion, this study reveals novel significance regarding the genetic characteristics and clinical value of ATP5B highlighted in predicting the outcome of breast cancer patients.
Collapse
Affiliation(s)
- Min Liu
- Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,The Graduate School, Tianjin Medical University, Tianjin, China
| | - Yuxuan Xu
- Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,The Graduate School, Tianjin Medical University, Tianjin, China
| | - Yaoyao Zhou
- Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,The Graduate School, Tianjin Medical University, Tianjin, China
| | - Ronggang Lang
- Department of Breast Cancer Pathology and Research Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhenyu Shi
- Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jing Zhao
- Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yuanyuan Meng
- Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China.,The Graduate School, Tianjin Medical University, Tianjin, China
| | - Li Bao
- Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
8
|
Samson JM, Ravindran Menon D, Smith DE, Baird E, Kitano T, Gao D, Tan AC, Fujita M. Clinical implications of ALDH1A1 and ALDH1A3 mRNA expression in melanoma subtypes. Chem Biol Interact 2019; 314:108822. [PMID: 31580832 DOI: 10.1016/j.cbi.2019.108822] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 08/23/2019] [Accepted: 09/12/2019] [Indexed: 12/15/2022]
Abstract
Aldehyde dehydrogenase (ALDH) activity is not only a valuable marker for cancer cells with stem-like features, but also plays a vital role in drug resistance and disease progression in many tumors including melanoma. However, the precise role of ALDH activity in patient prognosis remains unclear. In this study, using the Cancer Genome Atlas (TCGA) RNA-sequencing expression data, we analyzed gene expression of ALDH isozymes in melanoma tumors to define the expression patterns and the prognostic and predictive values of these enzymes. We found that ALDH1A1 and ALDH1A3 had both higher and broader expression ranges in melanoma patients, and that ALDH1A3 expression correlated with better overall survival in metastatic melanoma. Further, stratification of the TCGA cohorts by the mutational subtypes of melanoma specifically revealed that expression of ALDH1A3 correlated with better prognosis in metastatic BRAF-mutant melanoma while expression of ALDH1A1 correlated with better prognosis in BRAF wild-type melanoma. Gene set enrichment analysis (GSEA) of these cohorts identified upregulation in oxidative phosphorylation, adipogenesis, and fatty acid metabolism signaling in ALDH1Alo patients, suggesting BRAF/MEK inhibitor resistance in that subset of patients. On the other hand, GSEA of ALDH1A3hi cohorts revealed upregulation in glycolysis, hypoxia and angiogenesis, suggesting BRAF/MEK inhibitor sensitivity in that subset of patients. Gene expression analysis using pre-treatment tumor samples supports high ALDH1A3 expression before BRAF/MEK inhibitor treatment as predictive of better treatment response in BRAF-mutant melanoma patients. Our study provides evidence that high ALDH1A3 mRNA expression is not only a prognostic marker but also a predictive marker for BRAF/MEK inhibitor treatment response in BRAF-mutant metastatic melanoma patients.
Collapse
Affiliation(s)
- Jenny Mae Samson
- Department of Dermatology, University of Colorado Denver, Aurora, CO, 80045, United States
| | - Dinoop Ravindran Menon
- Department of Dermatology, University of Colorado Denver, Aurora, CO, 80045, United States
| | - Derek E Smith
- Department of Biostatistics & Informatics, University of Colorado Denver, Aurora, CO 80045, United States
| | - Erika Baird
- Department of Dermatology, University of Colorado Denver, Aurora, CO, 80045, United States
| | - Takayuki Kitano
- Department of Dermatology, University of Colorado Denver, Aurora, CO, 80045, United States; School of Medicine, University of the Ryukyus, Nishihara, Okinawa, 903-0215, Japan
| | - Dexiang Gao
- Department of Biostatistics & Informatics, University of Colorado Denver, Aurora, CO 80045, United States
| | - Aik-Choon Tan
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver, Aurora, CO, 80045, United States.
| | - Mayumi Fujita
- Department of Dermatology, University of Colorado Denver, Aurora, CO, 80045, United States; Denver VA Medical Center, Denver, CO, 80220, United States; Department of Immunology & Microbiology, University of Colorado Denver, Aurora, CO, 80045, United States.
| |
Collapse
|
9
|
Kwiatkowska-Borowczyk E, Czerwińska P, Mackiewicz J, Gryska K, Kazimierczak U, Tomela K, Przybyła A, Kozłowska AK, Galus Ł, Kwinta Ł, Dondajewska E, Gąbka-Buszek A, Żakowska M, Mackiewicz A. Whole cell melanoma vaccine genetically modified to stem cells like phenotype generates specific immune responses to ALDH1A1 and long-term survival in advanced melanoma patients. Oncoimmunology 2018; 7:e1509821. [PMID: 30377573 PMCID: PMC6205007 DOI: 10.1080/2162402x.2018.1509821] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/02/2018] [Accepted: 08/04/2018] [Indexed: 12/19/2022] Open
Abstract
Allogeneic whole cell gene modified therapeutic melanoma vaccine (AGI-101H) comprising of two melanoma cell lines transduced with cDNA encoding fusion protein composed of IL-6 linked with the soluble IL-6 receptor (sIL-6R), referred to as H6 was developed. H6 served as a molecular adjuvant, however, it has altered vaccine cells phenotype towards melanoma stem cells (MSC)-like with high activity of aldehyde dehydrogenase isoenzyme (ALDH1A1). AGI-101H was applied in advanced melanoma patients with non-resected and resected disease. In the adjuvant setting, it was combined with surgery in case of recurring metastases, which were surgically removed and vaccination continued. A significant fraction of AGI-101H treated melanoma patients is still alive (11–19 years). Out of 106 living patients, 39 were HLA-A2 positive and were the subject of the study. Immunization of melanoma patients resulted in the generation of cytotoxic CD8+ T cells specific for ALDH1A1, which were detected in circulation by HLA-A0201 MHC dextramers loaded with ALDH1A188-96(LLYKLADLI) peptide. Phenotypically they were central memory CD8+ T cells. Re-stimulation with ALDH1A188-96ex vivo resulted in IFN-γ secretion and cells degranulation. Following each vaccine dose administration, the number of ALDH1A1-CD8+ T cells increased in circulation and returned to the previous level until next dose injection (one month). ALDH1A1-CD8+ T cells were also found, however in the lower number than in vaccinated patients, in the circulation of untreated melanoma with stage IV but were not found in stage II or III and healthy donors. Specific anti-ALDH1 antibodies were present in treated patients. Long-term survival suggests immuno-targeting of MSC in treated patients.
Collapse
Affiliation(s)
- Eliza Kwiatkowska-Borowczyk
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland.,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan, Poland
| | - Patrycja Czerwińska
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland.,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan, Poland
| | - Jacek Mackiewicz
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan, Poland.,Department of Medical and Experimental Oncology, Heliodor Swiecicki University Hospital, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Gryska
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Urszula Kazimierczak
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Tomela
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Anna Przybyła
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Łukasz Galus
- Department of Medical and Experimental Oncology, Heliodor Swiecicki University Hospital, Poznan University of Medical Sciences, Poznan, Poland.,Department of Chemotherapy, Greater Poland Cancer Centre, Poznan, Poland
| | - Łukasz Kwinta
- Department of Chemotherapy, Greater Poland Cancer Centre, Poznan, Poland
| | - Ewelina Dondajewska
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Monika Żakowska
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Andrzej Mackiewicz
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland.,Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan, Poland
| |
Collapse
|