1
|
Belladelli F, De Cobelli F, Piccolo C, Cei F, Re C, Musso G, Rosiello G, Cignoli D, Santangelo A, Fallara G, Matloob R, Bertini R, Gusmini S, Brembilla G, Lucianò R, Tenace N, Salonia A, Briganti A, Montorsi F, Larcher A, Capitanio U. A machine learning-based analysis for the definition of an optimal renal biopsy for kidney cancer. Urol Oncol 2025; 43:270.e1-270.e8. [PMID: 39516081 DOI: 10.1016/j.urolonc.2024.10.020] [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] [Received: 08/06/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE Renal Tumor biopsy (RTB) can assist clinicians in determining the most suitable approach for treatment of renal cancer. However, RTB's limitations in accurately determining histology and grading have hindered its broader adoption and data on the concordance rate between RTB results and final pathology after surgery are unavailable. Therefore, we aimed to develop a machine learning algorithm to optimize RTB technique and to investigate the degree of concordance between RTB and surgical pathology reports. MATERIALS AND METHODS Within a prospectively maintained database, patients with indeterminate renal masses who underwent RTB at a single tertiary center were identified. We recorded and analyzed the approach (US vs. CT), the number of biopsy cores (NoC), and total core tissue length (LoC) to evaluate their impact on diagnostic outcomes. The K-Nearest Neighbors (KNN), a non-parametric supervised machine learning model, predicted the probability of obtaining pathological characterization and grading. In surgical patients, final pathology reports were compared with RTB results. RESULTS Overall, 197 patients underwent RTB. Overall, 89.8% (n=177) and 44.7% (n=88) of biopsies were informative in terms of histology and grading, respectively. The discrepancy rate between the pathology results from renal tissue biopsy (RTB) and the final pathology report following surgery was 3.6% (n=7) for histology and 5.0% (n=10) for grading. According to the machine learning model, a minimum of 2 cores providing at least 0.8 cm of total tissue should be obtained to achieve the best accuracy in characterizing the cancer. Alternatively, in cases of RTB with more than two cores, no specific minimum tissue threshold is required. CONCLUSIONS The discordance rates between RTB pathology and final surgical pathology are notably minimal. We defined an optimal renal biopsy strategy based on at least 2 cores and at least 0.8 cm of tissue or at least 3 cores and no minimum tissue threshold. PATIENTS SUMMARY RTB is a useful test for kidney cancer, but it's not always perfect. Our study shows that it usually matches up well with what doctors find during surgery. Using machine learning can make RTB even better by helping doctors know how many samples to take. This helps doctors treat kidney cancer more accurately.
Collapse
Affiliation(s)
- F Belladelli
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - F De Cobelli
- Department of Radiology, IRCCS San Raffaele Hospital, Milan, Italy
| | - C Piccolo
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - F Cei
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - C Re
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - G Musso
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - G Rosiello
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - D Cignoli
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - A Santangelo
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - G Fallara
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - R Matloob
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - R Bertini
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - S Gusmini
- Department of Radiology, IRCCS San Raffaele Hospital, Milan, Italy
| | - G Brembilla
- Department of Radiology, IRCCS San Raffaele Hospital, Milan, Italy
| | - R Lucianò
- Department of Pathology, IRCCS San Raffaele Hospital, Milan, Italy
| | - N Tenace
- Department of Pathology, IRCCS San Raffaele Hospital, Milan, Italy
| | - A Salonia
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - A Briganti
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - F Montorsi
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - A Larcher
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - U Capitanio
- URI - Urological Research Institute, Department of Urology, Division of Experimental Oncology, IRCCS San Raffaele Hospital, Milan, Italy; Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy.
| |
Collapse
|
2
|
Latcu SC, Bardan R, Cumpanas AA, Barbos V, Baderca F, Gaje PN, Ceausu RA, Comsa S, Dumitru CS, Dumache R, Cut TG, Lazureanu VE, Petrica L. Immunotherapy Applications for Thymine Dimers and WT1 Antigen in Renal Cancers: A Comparative Statistical Analysis. J Pers Med 2024; 14:557. [PMID: 38929778 PMCID: PMC11205122 DOI: 10.3390/jpm14060557] [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: 04/18/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Renal cell carcinoma (RCC) remains incurable in advanced stages. Biomarkers have proven to be quite useful in cancer therapeutics. Herein, we provide a comparative/integrative statistical analysis of seminal immunohistochemistry (IHC) findings for Wilms' Tumor 1 antigen (WT1) and thymine dimers (TDs), emerging as atypical, yet promising, potential biomarkers for RCCs. We assessed WT1/TD reactivity in adult RCC tumor cells, tumor microenvironment (TME), and tumor-adjacent healthy renal tissue (HRT). WT1 positivity was scarce and strictly nuclear in tumor cells, whereas TD-reactive tumor tissues were prevalent. We report statistically significant positive correlations between the density of reactive RCC cellularity and the intensity of nuclear staining for both biomarkers (WT1 - rho = 0.341, p-value = 0.036; TDs - rho = 0.379, p-value = 0.002). RCC stromal TME TD-positivity was much more frequent than WT1 reactivity, apparently proportional to that of the proper RCC cellularity and facilitated by extensive RCC inflammatory infiltration. TDs exhibited nuclear reactivity for most TME cell lines, while RCC TME WT1 expression was rare and inconsistent. In HRTs, TDs were entirely restricted to renal tubular cells, the likely cellular progenitor of most conventional RCC subtypes. In lieu of proper validation, these early findings have significant implications regarding the origins/biology of RCCs and may inform RCC therapeutics, both accounting for the high frequency of immunotherapy-permissive frameshift indels in RCCs, but also hinting at novel predictive clinical tools for WT1-targeted immunotherapy. Overall, the current study represents a meek yet hopefully significant step towards understanding the molecular biology and potential therapeutic targets of RCCs.
Collapse
Affiliation(s)
- Silviu Constantin Latcu
- Doctoral School, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania; (S.C.L.); (V.B.)
- Department XV, Discipline of Urology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Razvan Bardan
- Department XV, Discipline of Urology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Alin Adrian Cumpanas
- Department XV, Discipline of Urology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Vlad Barbos
- Doctoral School, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania; (S.C.L.); (V.B.)
| | - Flavia Baderca
- Department II of Microscopic Morphology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania; (F.B.); (P.N.G.); (R.A.C.); (S.C.); (C.-S.D.)
- Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Pusa Nela Gaje
- Department II of Microscopic Morphology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania; (F.B.); (P.N.G.); (R.A.C.); (S.C.); (C.-S.D.)
- Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Raluca Amalia Ceausu
- Department II of Microscopic Morphology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania; (F.B.); (P.N.G.); (R.A.C.); (S.C.); (C.-S.D.)
- Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Serban Comsa
- Department II of Microscopic Morphology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania; (F.B.); (P.N.G.); (R.A.C.); (S.C.); (C.-S.D.)
- Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Cristina-Stefania Dumitru
- Department II of Microscopic Morphology, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania; (F.B.); (P.N.G.); (R.A.C.); (S.C.); (C.-S.D.)
- Angiogenesis Research Center, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania
| | - Raluca Dumache
- Department VIII, Discipline of Forensic Medicine, Bioethics, Deontology and Medical Law, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania;
- Center for Ethics in Human Genetic Identifications, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Talida Georgiana Cut
- Center for Ethics in Human Genetic Identifications, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania;
- Department XIII, Discipline of Infectious Diseases, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Voichita Elena Lazureanu
- Department XIII, Discipline of Infectious Diseases, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Ligia Petrica
- Department of Internal Medicine II, Division of Nephrology, Victor Babes University of Medicine and Pharmacy Timisoara, County Emergency Hospital Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania;
- Centre for Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, Victor Babes University of Medicine and Pharmacy Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania
| |
Collapse
|
3
|
Buhas BA, Toma V, Beauval JB, Andras I, Couți R, Muntean LAM, Coman RT, Maghiar TA, Știufiuc RI, Lucaciu CM, Crisan N. Label-Free SERS of Urine Components: A Powerful Tool for Discriminating Renal Cell Carcinoma through Multivariate Analysis and Machine Learning Techniques. Int J Mol Sci 2024; 25:3891. [PMID: 38612705 PMCID: PMC11011951 DOI: 10.3390/ijms25073891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 03/29/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
The advent of Surface-Enhanced Raman Scattering (SERS) has enabled the exploration and detection of small molecules, particularly in biological fluids such as serum, blood plasma, urine, saliva, and tears. SERS has been proposed as a simple diagnostic technique for various diseases, including cancer. Renal cell carcinoma (RCC) ranks as the sixth most commonly diagnosed cancer in men and is often asymptomatic, with detection occurring incidentally. The onset of symptoms typically aligns with advanced disease, aggressive histology, and unfavorable prognosis, and therefore new methods for an early diagnosis are needed. In this study, we investigated the utility of label-free SERS in urine, coupled with two multivariate analysis approaches: Principal Component Analysis combined with Linear Discriminant Analysis (PCA-LDA) and Support Vector Machine (SVM), to discriminate between 50 RCC patients and 44 healthy donors. Employing LDA-PCA, we achieved a discrimination accuracy of 100% using 13 principal components, and an 88% accuracy in discriminating between different RCC stages. The SVM approach yielded a training accuracy of 100%, a validation accuracy of 99% for discriminating between RCC and controls, and an 80% accuracy for discriminating between stages. The comparative analysis of raw and normalized SERS spectral data shows that while raw data disclose relative concentration variations in urine metabolites between the two classes, the normalization of spectral data significantly improves the accuracy of discrimination. Moreover, the selection of principal components with markedly distinct scores between the two classes serves to alleviate overfitting risks and reduces the number of components employed for discrimination. We obtained the accuracy of the discrimination between the RCC patients cases and healthy donors of 90% for three PCs and a linear discrimination function, and a 88% accuracy of discrimination between stages using six PCs, mitigating practically the risk of overfitting and increasing the robustness of our analysis. Our findings underscore the potential of label-free SERS of urine in conjunction with chemometrics for non-invasive and early RCC detection.
Collapse
Affiliation(s)
- Bogdan Adrian Buhas
- Department of Urology, La Croix du Sud Hospital, 52 Chemin de Ribaute St., 31130 Quint Fonsegrives, France; (B.A.B.); (J.-B.B.)
- Department of Urology, Clinical Municipal Hospital, 11 Tabacarilor St., 400139 Cluj-Napoca, Romania; (I.A.); (N.C.)
- Faculty of Medicine and Pharmacy, University of Oradea, 1 Universitatii St., 410087 Oradea, Romania; (R.C.); (T.A.M.)
| | - Valentin Toma
- Department of Nanobiophysics, MedFuture Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 4-6 Pasteur St., 400337 Cluj-Napoca, Romania;
| | - Jean-Baptiste Beauval
- Department of Urology, La Croix du Sud Hospital, 52 Chemin de Ribaute St., 31130 Quint Fonsegrives, France; (B.A.B.); (J.-B.B.)
| | - Iulia Andras
- Department of Urology, Clinical Municipal Hospital, 11 Tabacarilor St., 400139 Cluj-Napoca, Romania; (I.A.); (N.C.)
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes St., 400347 Cluj-Napoca, Romania
| | - Răzvan Couți
- Faculty of Medicine and Pharmacy, University of Oradea, 1 Universitatii St., 410087 Oradea, Romania; (R.C.); (T.A.M.)
| | - Lucia Ana-Maria Muntean
- Department of Medical Education, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes St., 400347 Cluj-Napoca, Romania;
| | - Radu-Tudor Coman
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes St., 400347 Cluj-Napoca, Romania
| | - Teodor Andrei Maghiar
- Faculty of Medicine and Pharmacy, University of Oradea, 1 Universitatii St., 410087 Oradea, Romania; (R.C.); (T.A.M.)
| | - Rareș-Ionuț Știufiuc
- Department of Nanobiophysics, MedFuture Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 4-6 Pasteur St., 400337 Cluj-Napoca, Romania;
- Department of Pharmaceutical Physics–Biophysics, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur St., 400349 Cluj-Napoca, Romania
- Nanotechnology Laboratory, TRANSCEND Research Center, Regional Institute of Oncology, 700483 Iași, Romania
| | - Constantin Mihai Lucaciu
- Department of Pharmaceutical Physics–Biophysics, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 6 Pasteur St., 400349 Cluj-Napoca, Romania
| | - Nicolae Crisan
- Department of Urology, Clinical Municipal Hospital, 11 Tabacarilor St., 400139 Cluj-Napoca, Romania; (I.A.); (N.C.)
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 8 Victor Babes St., 400347 Cluj-Napoca, Romania
| |
Collapse
|
4
|
Azawi N, Mosholt KSS, Fryd ND, Lund L, Brignone JI, Hvid N, Wulf-Johansson H, Pedersen OBV, Saekmose SG, Dabestani S. suPARnostic: an advanced predictive tool for detecting recurrence in renal cell carcinoma. BMC Urol 2023; 23:168. [PMID: 37875832 PMCID: PMC10594785 DOI: 10.1186/s12894-023-01337-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/06/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Plasma soluble urokinase-type Plasminogen Activator Receptor (suPAR) predicts disease aggressiveness in renal cell carcinoma (ccRCC), but its prognostic accuracy has not been investigated. To investigate the prognostic accuracy of preoperative plasma suPAR in patients who received curative treatment for initially localized ccRCC. METHODS We retrospectively analyzed plasma samples stored in the Danish National Biobank between 2010 and 2015 from 235 patients with ccRCC at any stage. Relationships with outcome analyzed using univariate and multiple logistic Cox regression analysis. RESULTS There were 235 patients with ccRCC. The median follow-up period was 7.7 years. In univariate analysis suPAR ≥ 6 ng/mL was significantly associated with overall survival (OS) and recurrence-free survival (RFS). Patients with elevated suPAR were more likely to recur, with a Hazard Ratio (HR) of 2.3 for RFS. In multiple logistic regression, suPAR ≥ 6 ng/mL remained a negative predictor of OS and RFS. Limitations include retrospective study design, wide confidence intervals, and tumor subtype heterogeneity bias. CONCLUSIONS ccRCC patients with high plasma suPAR concentrations are at an elevated risk of disease recurrence and see lower OS. suPAR is a promising surveillance tool to more precisely follow up with ccRCC patients and detect future recurrences. In this study, we showed that new type of liquid marker in blood plasma, called suPAR, is associated to a higher risk of kidney cancer recurrence when elevated above 6ng/mL. We also showed suPAR to independently be able to predict patients overall and recurrence free survival in patient with any stage of kidney cancer.
Collapse
Affiliation(s)
- Nessn Azawi
- Department of Urology, Zealand University Hospital, Sygehusvej 6, Roskilde, 4000, Denmark.
- Institute of Clinical Medicine, University of Copenhagen, Norregade 10, copenhagen, Copenhagen, 1165, Denmark.
| | | | - Nathalie Demuth Fryd
- Deparment of Urology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Aarhus, Denmark
| | - Lars Lund
- Deparment of Urology, Odense University Hospital, J. B. Winslows Vej 4, Odense, 5000, Denmark
| | - Juan Ignacio Brignone
- Department of Urology, Aalborg University hospital, Hobrovej 18-22, Aalborg, Aalborg, 9100, Denmark
| | - Nanna Hvid
- Deparment of Urology, Odense University Hospital, J. B. Winslows Vej 4, Odense, 5000, Denmark
| | - Helle Wulf-Johansson
- Department of Clinical Immunology, Naestved Hospital, Ringstedgade 61, Naestved, Naestved, 4700, Denmark
| | | | - Susanne Gjørup Saekmose
- Department of Clinical Immunology, Naestved Hospital, Ringstedgade 61, Naestved, Naestved, 4700, Denmark
| | - Saeed Dabestani
- Department of Urology, Kristianstad Central Hospital, Region Skane, Kristianstad, Sweden
- Department of Translational Medicine, Division of Urological Cancers, Lund University, Lund, Sweden
| |
Collapse
|
5
|
Li Y, Hashikawa K, Ebisawa K, Kambe M, Higuchi S, Kamei Y. <Editors' Choice> Supernatant from activated omentum accelerates wound healing in diabetic mice wound model. NAGOYA JOURNAL OF MEDICAL SCIENCE 2023; 85:528-541. [PMID: 37829482 PMCID: PMC10565575 DOI: 10.18999/nagjms.85.3.528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/04/2022] [Indexed: 10/14/2023]
Abstract
Diabetic wounds are considered one of the most frequent and severe complications of diabetes mellitus. Recently, the omentum has been used in diabetic wound healing because of its tissue repair properties. The activated omentum is richer in growth factors than the inactivated, thereby contributing to the wound healing process. To further investigate the effect of activated omentum conditioned medium (aOCM) on diabetic wound healing, we injected supernatant from aOCM, saline-OCM (sOCM), inactivated-OCM (iOCM), and medium (M) subcutaneously upon creation of a cutaneous wound healing model in diabetic mice. Wound area (%) was evaluated on days 0, 3, 5, 7, 9, 11, 14, 21, and 28 post-operation. At 9 and 28 d post-operation, skin tissue was harvested and assessed for gross observation, neovascularization, peripheral nerve fiber regeneration, and collagen deposition. We observed that aOCM enhanced the wound repair process, with significant acceleration of epidermal and collagen deposition in the surgical lesion on day 9. Additionally, aOCM displayed marked efficiency in neovascularization and peripheral nerve regeneration during wound healing. Thus, aOCM administration exerts a positive influence on the diabetic mouse model, which can be employed as a new therapy for diabetic wounds.
Collapse
Affiliation(s)
- Yu Li
- Department of Plastic and Reconstructive Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazunobu Hashikawa
- Department of Plastic and Reconstructive Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Katsumi Ebisawa
- Department of Plastic and Reconstructive Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Miki Kambe
- Department of Plastic and Reconstructive Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinichi Higuchi
- Department of Plastic and Reconstructive Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuzuru Kamei
- Department of Plastic and Reconstructive Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
6
|
Contemporary Clinical Definitions, Differential Diagnosis, and Novel Predictive Tools for Renal Cell Carcinoma. Biomedicines 2022; 10:biomedicines10112926. [PMID: 36428491 PMCID: PMC9687297 DOI: 10.3390/biomedicines10112926] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/26/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Despite significant progress regarding clinical detection/imaging evaluation modalities and genetic/molecular characterization of pathogenesis, advanced renal cell carcinoma (RCC) remains an incurable disease and overall RCC mortality has been steadily rising for decades. Concomitantly, clinical definitions have been greatly nuanced and refined. RCCs are currently viewed as a heterogeneous series of cancers, with the same anatomical origin, but fundamentally different metabolisms and clinical behaviors. Thus, RCC pathological diagnosis/subtyping guidelines have become increasingly intricate and cumbersome, routinely requiring ancillary studies, mainly immunohistochemistry. Meanwhile, RCC-associated-antigen targeted systemic therapy has been greatly diversified and emerging, novel clinical applications for RCC immunotherapy have already reported significant survival benefits, at least in the adjuvant setting. Even so, systemically disseminated RCCs still associate very poor clinical outcomes, with currently available therapeutic modalities only being able to prolong survival. In lack of a definitive cure for advanced RCCs, integration of the amounting scientific knowledge regarding RCC pathogenesis into RCC clinical management has been paramount for improving patient outcomes. The current review aims to offer an integrative perspective regarding contemporary RCC clinical definitions, proper RCC clinical work-up at initial diagnosis (semiology and multimodal imaging), RCC pathological evaluation, differential diagnosis/subtyping protocols, and novel clinical tools for RCC screening, risk stratification and therapeutic response prediction.
Collapse
|
7
|
Panels of mRNAs and miRNAs for decoding molecular mechanisms of Renal Cell Carcinoma (RCC) subtypes utilizing Artificial Intelligence approaches. Sci Rep 2022; 12:16393. [PMID: 36180558 PMCID: PMC9525704 DOI: 10.1038/s41598-022-20783-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 09/19/2022] [Indexed: 11/12/2022] Open
Abstract
Renal Cell Carcinoma (RCC) encompasses three histological subtypes, including clear cell RCC (KIRC), papillary RCC (KIRP), and chromophobe RCC (KICH) each of which has different clinical courses, genetic/epigenetic drivers, and therapeutic responses. This study aimed to identify the significant mRNAs and microRNA panels involved in the pathogenesis of RCC subtypes. The mRNA and microRNA transcripts profile were obtained from The Cancer Genome Atlas (TCGA), which were included 611 ccRCC patients, 321 pRCC patients, and 89 chRCC patients for mRNA data and 616 patients in the ccRCC subtype, 326 patients in the pRCC subtype, and 91 patients in the chRCC for miRNA data, respectively. To identify mRNAs and miRNAs, feature selection based on filter and graph algorithms was applied. Then, a deep model was used to classify the subtypes of the RCC. Finally, an association rule mining algorithm was used to disclose features with significant roles to trigger molecular mechanisms to cause RCC subtypes. Panels of 77 mRNAs and 73 miRNAs could discriminate the KIRC, KIRP, and KICH subtypes from each other with 92% (F1-score ≥ 0.9, AUC ≥ 0.89) and 95% accuracy (F1-score ≥ 0.93, AUC ≥ 0.95), respectively. The Association Rule Mining analysis could identify miR-28 (repeat count = 2642) and CSN7A (repeat count = 5794) along with the miR-125a (repeat count = 2591) and NMD3 (repeat count = 2306) with the highest repeat counts, in the KIRC and KIRP rules, respectively. This study found new panels of mRNAs and miRNAs to distinguish among RCC subtypes, which were able to provide new insights into the underlying responsible mechanisms for the initiation and progression of KIRC and KIRP. The proposed mRNA and miRNA panels have a high potential to be as biomarkers of RCC subtypes and should be examined in future clinical studies.
Collapse
|
8
|
Kim RD, Kovari BP, Martinez M, Xie H, Sahin IH, Mehta R, Strosberg J, Imanirad I, Ghayouri M, Kim YC, Kim DW. A phase I/Ib study of regorafenib and nivolumab in mismatch repair proficient advanced refractory colorectal cancer. Eur J Cancer 2022; 169:93-102. [DOI: 10.1016/j.ejca.2022.03.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/07/2022] [Accepted: 03/19/2022] [Indexed: 12/17/2022]
|
9
|
Abstract
Melanoma is a relentless type of skin cancer which involves myriad signaling pathways which regulate many cellular processes. This makes melanoma difficult to treat, especially when identified late. At present, therapeutics include chemotherapy, surgical resection, biochemotherapy, immunotherapy, photodynamic and targeted approaches. These interventions are usually administered as either a single-drug or in combination, based on tumor location, stage, and patients' overall health condition. However, treatment efficacy generally decreases as patients develop treatment resistance. Genetic profiling of melanocytes and the discovery of novel molecular factors involved in the pathogenesis of melanoma have helped to identify new therapeutic targets. In this literature review, we examine several newly approved therapies, and briefly describe several therapies being assessed for melanoma. The goal is to provide a comprehensive overview of recent developments and to consider future directions in the field of melanoma.
Collapse
Affiliation(s)
- Pavan Kumar Dhanyamraju
- Department of Pediatrics and Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Pavan Kumar Dhanyamraju, Department of Pediatrics and Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA17033, USA. Tel: +1-6096474712, E-mail:
| | - Trupti N. Patel
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Vellore, Tamil Nadu 632014, India
| |
Collapse
|
10
|
Fasanella D, Antonaci A, Esperto F, Scarpa RM, Ferro M, Schips L, Marchioni M. Potential prognostic value of miRNAs as biomarker for progression and recurrence after nephrectomy in renal cell carcinoma: a literature review. Diagnosis (Berl) 2021; 9:157-165. [PMID: 34674417 DOI: 10.1515/dx-2021-0080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 10/06/2021] [Indexed: 01/01/2023]
Abstract
OBJECTIVES We provide a systematic literature review on tissue miRNAs in patients with RCC to evaluate and summarize their usefulness as prognostic markers. We undertook a systematic search for articles in English using the PubMed-Medline database from January 2010 to December 2020. Studies were identified and selected according to the PRISMA criteria and the PICO methodology. The population consisted of RCC patients undergoing nephrectomy and the main outcome of interest was recurrence-free survival (RFS). Only studies providing hazard ratios (HRs) from multivariate or univariate analyzes with corresponding 95% confidence intervals (CI) and/or area under the curve (AUC) were considered. CONTENT All nine included studies (1,541 patients) analyzed the relationship between tissue miRNA expression levels (up or downregulated) and RFS. Some of these found that the methylation status of miR-9-1, miR-9-3 and miR-124 was associated with a high risk of relapse. Moreover, miR-200b overexpression was associated with OS. MiR-210 overexpression indicated a shorter OS than those who were miR-210 negative. Finally, patients with high miR-125b expression had shorter CSS than those with low expression; similarly, patients with low miR-126 expression also had shorter CSS time. SUMMARY AND OUTLOOK Several studies tested the usefulness of specific miRNAs to predict RCC recurrence. Some of them showed a fair accuracy and strong relationship between specific miRNA over or under-expression and survival outcomes. However, results from these studies are preliminary and miRNAs use in routine clinical practice is still far to come.
Collapse
Affiliation(s)
- Daniela Fasanella
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti, Urology Unit, SS Annunziata Hospital, Chieti, Italy
| | - Alessio Antonaci
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti, Urology Unit, SS Annunziata Hospital, Chieti, Italy
| | - Francesco Esperto
- Department of Urology, Campus Biomedico University of Rome, Rome, Italy
| | - Roberto M Scarpa
- Department of Urology, Campus Biomedico University of Rome, Rome, Italy
| | - Matteo Ferro
- Division of Urology, European Institute of Oncology-IRCCS, Milan, Italy
| | - Luigi Schips
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti, Urology Unit, SS Annunziata Hospital, Chieti, Italy
| | - Michele Marchioni
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti, Urology Unit, SS Annunziata Hospital, Chieti, Italy
| |
Collapse
|
11
|
Butner JD, Wang Z, Elganainy D, Al Feghali KA, Plodinec M, Calin GA, Dogra P, Nizzero S, Ruiz-Ramírez J, Martin GV, Tawbi HA, Chung C, Koay EJ, Welsh JW, Hong DS, Cristini V. A mathematical model for the quantification of a patient's sensitivity to checkpoint inhibitors and long-term tumour burden. Nat Biomed Eng 2021; 5:297-308. [PMID: 33398132 PMCID: PMC8669771 DOI: 10.1038/s41551-020-00662-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/14/2020] [Indexed: 02/06/2023]
Abstract
A large proportion of patients with cancer are unresponsive to treatment with immune checkpoint blockade and other immunotherapies. Here, we report a mathematical model of the time course of tumour responses to immune checkpoint inhibitors. The model takes into account intrinsic tumour growth rates, the rates of immune activation and of tumour-immune cell interactions, and the efficacy of immune-mediated tumour killing. For 124 patients, four cancer types and two immunotherapy agents, the model reliably described the immune responses and final tumour burden across all different cancers and drug combinations examined. In validation cohorts from four clinical trials of checkpoint inhibitors (with a total of 177 patients), the model accurately stratified the patients according to reduced or increased long-term tumour burden. We also provide model-derived quantitative measures of treatment sensitivity for specific drug-cancer combinations. The model can be used to predict responses to therapy and to quantify specific drug-cancer sensitivities in individual patients.
Collapse
Affiliation(s)
- Joseph D Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Dalia Elganainy
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Karine A Al Feghali
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marija Plodinec
- Biozentrum and the Swiss Nanoscience Institute, University of Basel, Basel, Switzerland
| | - George A Calin
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Sara Nizzero
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Javier Ruiz-Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Geoffrey V Martin
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hussein A Tawbi
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caroline Chung
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eugene J Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James W Welsh
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
12
|
Radovanović M, Džamić Z. Autophagy and renal cell carcinoma: What do we know so far? MEDICINSKI PODMLADAK 2021. [DOI: 10.5937/mp72-31557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Renal cell carcinoma (RCC) is the most common type of kidney tumor in adults, accounting for approximately 90% of kidney malignances, occurring usually between the ages of 60 and 70. The 5-year overall survival rate for all RCC types is 49%. Since RCCs are resistant to numeorus different radio and chemotherapeutics that act via apoptosis induction, the development of new approaches to RCC treatment is still in the focus of modern urology. In particular, in recent years, autophagy in RCC has been widely studied as a mechanism of cell extinction through which tumor cells can overcome resistance to apoptosis activation therapy. Autophagy is often referred to as a double-edged sword because it can be a process that allows cells of cancer to survive and, on the other hand and under other conditions, it can be a cell dying mechanism, independent or closely related to other cell death modalities, like apoptosis and necrosis. The central role in the tempering of the process of autophagy, in general, belongs to the mTOR complex (mammalian target of rapamycin), which integrates numerous signals that affect autophagy, such as growth factors, nutrients, various stressors and the energy status of the cell. In RCC, the most important is PI3K/AKT/mTOR signaling pathway, since activation of this signaling leads to survival of tumor cells through mTOR activation and thus, autophagy inhibition. Up to now, it was found that autophagy markers such as Beclin-1 and LC3-II can be considered as prognostic markers for RCC since the high level of Beclin-1 was detected in tissues and cells of RCC (A498 and ACHN cell lines) and that tumor cell mobility is promoted by the up-regulated expression of LC3. Therefore, a progress in RCC therapy can be expected from the development and synthesis of specific compounds targeting autophagy, as well as the therapy based on their combination.
Collapse
|
13
|
Liu X, Xu Y, Li Y, Pan Y, Zhao S, Hou Y. Ferumoxytol-β-glucan Inhibits Melanoma Growth via Interacting with Dectin-1 to Polarize Macrophages into M1 Phenotype. Int J Med Sci 2021; 18:3125-3139. [PMID: 34400883 PMCID: PMC8364471 DOI: 10.7150/ijms.61525] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 06/14/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Regulating the polarization of macrophages to antitumor M1 macrophages is a promising strategy for overcoming the immunosuppression of the tumor microenvironment for cancer therapy. Ferumoxytol (FMT) can not only serve as a drug deliver agent but also exerts anti-tumor activity. β-glucan has immuno-modulating properties to prevent tumor growth. Thus, a nanocomposite of FMT surface-coated with β-glucan (FMT-β-glucan) was prepared to explore its effect on tumor suppression. Methods: Male B16F10 melanoma mouse model was established to explore the antitumor effect of FMT-β-glucan. The viability and apoptotic rates of B16F10 cells were detected by cell counting kit-8 and Annexin-V/PI experiments. The levels of M1 markers were quantified by quantitative reverse transcription-polymerase chain reaction and enzyme linked immunosorbent assay. Phagocytic activity and intracellular reactive oxygen species (ROS) in macrophages were evaluated by the neutral red uptake assay and flow cytometry, respectively. Small interfering RNA (siRNA) transfection was applied to knock down the Dectin-1 gene in RAW 264.7 cells. Results: FMT-β-glucan suppressed tumor growth to a greater extent and induced higher infiltration of M1 macrophages than the combination of FMT and β-glucan (FMT+β-glucan) in vivo. In vitro, supernatant from FMT-β-glucan-treated RAW 264.7 cells led to lower cell viability and induced more apoptosis of B16F10 cells than that from the FMT+β-glucan group. Moreover, FMT-β-glucan boosted the expression of M1 type markers, and increased phagocytic activity and ROS in RAW 264.7 cells. Further research indicated that FMT-β-glucan treatment promoted the level of Dectin-1 on the surface of RAW 264.7 cells and that knockdown of Dectin-1 abrogated the phosphorylation levels of several components in MAPK and NF-κB signaling. Conclusion: The nanocomposite FMT-β-glucan suppressed melanoma growth by inducing the M1 macrophage-activated tumor microenvironment.
Collapse
Affiliation(s)
- Xinghan Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Yujun Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Yi Li
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Yuchen Pan
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
| | - Shuli Zhao
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
- ✉ Corresponding authors: Yayi Hou, The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China. Tel.: +86-25-8968-8441; Fax: +86-25-8968-8441. E-mail: ; Shuli Zhao, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China. E-mail:
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing 210093, China
- ✉ Corresponding authors: Yayi Hou, The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing 210093, China. Tel.: +86-25-8968-8441; Fax: +86-25-8968-8441. E-mail: ; Shuli Zhao, General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China. E-mail:
| |
Collapse
|
14
|
Khattak MA, Abed A, Reid AL, McEvoy AC, Millward M, Ziman M, Gray ES. Role of Serum Vascular Endothelial Growth Factor (VEGF) as a Potential Biomarker of Response to Immune Checkpoint Inhibitor Therapy in Advanced Melanoma: Results of a Pilot Study. Front Oncol 2020; 10:1041. [PMID: 32695680 PMCID: PMC7338663 DOI: 10.3389/fonc.2020.01041] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 05/26/2020] [Indexed: 01/21/2023] Open
Abstract
Background: The development of biomarkers predictive of response to immune checkpoint inhibitor (ICI) therapies in advanced melanoma is an area of great interest in oncology. Our study evaluated the potential role of serum vascular endothelial growth factor (VEGF) as a predictive biomarker of clinical benefit and response to treatment with ICIs. Methods: Pre-treatment peripheral blood samples were obtained from advanced melanoma patients undergoing ICI therapy as monotherapy or in combination at two tertiary care hospitals in Western Australia. Serum VEGF levels were correlated with response to therapy and survival outcomes. Results: Serum VEGF samples were collected from a total of 130 patients treated with ICI therapy (pembrolizumab 73, ipilimumab 15, and ipilimumab/nivolumab combination 42). Median serum VEGF level was significantly higher in the non-responders (82.15 pg/mL) vs. responders (60.40 pg/mL) in the ipilimumab monotherapy cohort (P < 0.0352). However, no difference was seen in VEGF levels between non-responders and responders in pembrolizumab and ipilimumab/nivolumab treated patients. Conclusions: The results of our study confirm previous observations that that high pre-treatment serum VEGF levels in advanced melanoma patients may predict poor response to ipilimumab. However, serum VEGF is not predictive of outcome in patients treated with anti-PD-1 agents alone or in combination with ipilimumab.
Collapse
Affiliation(s)
- Muhammad A Khattak
- Department of Medical Oncology, Fiona Stanley Hospital, Murdoch, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia.,School of Medicine, University of Western Australia, Crawley, WA, Australia
| | - Afaf Abed
- Department of Medical Oncology, Fiona Stanley Hospital, Murdoch, WA, Australia.,School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Anna L Reid
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Ashleigh C McEvoy
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Michael Millward
- School of Medicine, University of Western Australia, Crawley, WA, Australia.,Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Melanie Ziman
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| | - Elin S Gray
- School of Medical and Health Sciences, Edith Cowan University, Perth, WA, Australia
| |
Collapse
|
15
|
Walsh RJ, Soo RA. Resistance to immune checkpoint inhibitors in non-small cell lung cancer: biomarkers and therapeutic strategies. Ther Adv Med Oncol 2020; 12:1758835920937902. [PMID: 32670423 PMCID: PMC7339077 DOI: 10.1177/1758835920937902] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 06/04/2020] [Indexed: 12/12/2022] Open
Abstract
The treatment landscape for patients with advanced non-small cell lung cancer has
evolved greatly with the advent of immune checkpoint inhibitors. However, many
patients do not derive benefit from checkpoint blockade, developing either
primary or secondary resistance, highlighting a need for alternative approaches
to modulate immune function. In this review, we highlight the absence of a
common definition of primary and secondary resistance and summarize their
frequency and clinical characteristics. Furthermore, we provide an overview of
the biomarkers and mechanisms of resistance involving the tumor, the tumor
microenvironment and the host, and suggest treatment strategies to overcome
these mechanisms and improve clinical outcomes.
Collapse
Affiliation(s)
- Robert J. Walsh
- Department of Haematology–Oncology, National
University Cancer Institute Singapore, Singapore
| | | |
Collapse
|
16
|
Acebes-Fernández V, Landeira-Viñuela A, Juanes-Velasco P, Hernández AP, Otazo-Perez A, Manzano-Román R, Gongora R, Fuentes M. Nanomedicine and Onco-Immunotherapy: From the Bench to Bedside to Biomarkers. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1274. [PMID: 32610601 PMCID: PMC7407304 DOI: 10.3390/nano10071274] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/16/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022]
Abstract
The broad relationship between the immune system and cancer is opening a new hallmark to explore for nanomedicine. Here, all the common and synergy points between both areas are reviewed and described, and the recent approaches which show the progress from the bench to the beside to biomarkers developed in nanomedicine and onco-immunotherapy.
Collapse
Affiliation(s)
- Vanessa Acebes-Fernández
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Alicia Landeira-Viñuela
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Pablo Juanes-Velasco
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Angela-Patricia Hernández
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Andrea Otazo-Perez
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Raúl Manzano-Román
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain;
| | - Rafael Gongora
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Manuel Fuentes
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain;
| |
Collapse
|
17
|
Arora S, Velichinskii R, Lesh RW, Ali U, Kubiak M, Bansal P, Borghaei H, Edelman MJ, Boumber Y. Existing and Emerging Biomarkers for Immune Checkpoint Immunotherapy in Solid Tumors. Adv Ther 2019; 36:2638-2678. [PMID: 31410780 PMCID: PMC6778545 DOI: 10.1007/s12325-019-01051-z] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Indexed: 02/06/2023]
Abstract
In the last few years, immunotherapy has transformed the way we treat solid tumors, including melanoma, lung, head neck, breast, renal, and bladder cancers. Durable responses and long-term survival benefit has been experienced by many cancer patients, with favorable toxicity profiles of immunotherapeutic agents relative to chemotherapy. Cures have become possible in some patients with metastatic disease. Additional approvals of immunotherapy drugs and in combination with other agents are anticipated in the near future. Multiple additional immunotherapy drugs are in earlier stages of clinical development, and their testing in additional tumor types is under way. Despite considerable early success and relatively fewer side effects, the majority of cancer patients do not respond to checkpoint inhibitors. Additionally, while the drugs are generally well tolerated, there is still the potential for significant, unpredictable and even fatal toxicity with these agents. Improved biomarkers may help to better select patients who are more likely to respond to these drugs. Two key biologically important predictive tissue biomarkers, specifically, PD-L1 and mismatch repair deficiency, have been FDA-approved in conjunction with the checkpoint inhibitor, pembrolizumab. Tumor mutation burden, another promising biomarker, is emerging in several tumor types, and may also soon receive approval. Finally, several other tissue and liquid biomarkers are emerging that could help guide single-agent immunotherapy and in combination with other agents. Of these, one promising investigational biomarker is alteration or deficiency in DNA damage response (DDR) pathways, with altered DDR observed in a broad spectrum of tumors. Here, we provide a critical overview of current, emerging, and investigational biomarkers in the context of response to immunotherapy in solid tumors.
Collapse
Affiliation(s)
- Sanjeevani Arora
- Program in Cancer Prevention and Control, Fox Chase Cancer Center, Philadelphia, PA, USA.
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Rodion Velichinskii
- Program in Cancer Prevention and Control, Fox Chase Cancer Center, Philadelphia, PA, USA
- Department of Molecular Biology and Medical Biotechnology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Randy W Lesh
- Program in Cancer Prevention and Control, Fox Chase Cancer Center, Philadelphia, PA, USA
- Department of Radiation Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
- Geisinger Commonwealth School of Medicine, Scranton, PA, USA
| | - Usman Ali
- Division of Hospital Medicine, Department of Medicine, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Michal Kubiak
- Internal Medicine Residency Program, Centegra Health System, McHenry Hospital and Rosalind Franklin University, Mchenry, IL, USA
| | | | - Hossein Borghaei
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Martin J Edelman
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Yanis Boumber
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA.
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.
| |
Collapse
|
18
|
Unraveling the crosstalk between melanoma and immune cells in the tumor microenvironment. Semin Cancer Biol 2019; 59:236-250. [PMID: 31404607 DOI: 10.1016/j.semcancer.2019.08.002] [Citation(s) in RCA: 222] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/10/2019] [Accepted: 08/04/2019] [Indexed: 12/12/2022]
Abstract
Cutaneous melanoma is the most common skin cancer with an incidence that has been rapidly increasing in the past decades. Melanomas are among the most immunogenic tumors and, as such, have the greatest potential to respond favorably to immunotherapy. However, like many cancers, melanomas acquire various suppressive mechanisms, which generally act in concert, to escape innate and adaptive immune detection and destruction. Intense research into the cellular and molecular events associated with melanomagenesis, which ultimately lead to immune suppression, has resulted in the discovery of new therapeutic targets and synergistic combinations of immunotherapy, targeted therapy and chemotherapy. Tremendous effort to determine efficacy of single and combination therapies in pre-clinical and clinical phase I-III trials has led to FDA-approval of several immunotherapeutic agents that could potentially be beneficial for aggressive, highly refractory, advanced and metastatic melanomas. The increasing availability of approved combination therapies for melanoma and more rapid assessment of patient tumors has increased the feasibility of personalized treatment to overcome patient and tumor heterogeneity and to achieve greater clinical benefit. Here, we review the evolution of the immune system during melanomagenesis, mechanisms exploited by melanoma to suppress anti-tumor immunity and methods that have been developed to restore immunity. We emphasize that an effective therapeutic strategy will require coordinate activation of tumor-specific immunity as well as increased recognition and accessibility of melanoma cells in primary tumors and distal metastases. This review integrates available knowledge on melanoma-specific immunity, molecular signaling pathways and molecular targeting strategies that could be utilized to envision therapeutics with broader application and greater efficacy for early stage and advanced metastatic melanoma.
Collapse
|
19
|
Caporali S, Amaro A, Levati L, Alvino E, Lacal PM, Mastroeni S, Ruffini F, Bonmassar L, Antonini Cappellini GC, Felli N, Carè A, Pfeffer U, D'Atri S. miR-126-3p down-regulation contributes to dabrafenib acquired resistance in melanoma by up-regulating ADAM9 and VEGF-A. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:272. [PMID: 31227006 PMCID: PMC6588909 DOI: 10.1186/s13046-019-1238-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 05/21/2019] [Indexed: 02/06/2023]
Abstract
Background Development of resistance to inhibitors of BRAF (BRAFi) and MEK (MEKi) remains a great challenge for targeted therapy in patients with BRAF-mutant melanoma. Here, we explored the role of miRNAs in melanoma acquired resistance to BRAFi. Methods miRNA expression in two BRAF-mutant melanoma cell lines and their dabrafenib-resistant sublines was determined using Affymetrix GeneChip® miRNA 3.1 microarrays and/or qRT-PCR. The effects of miR-126-3p re-expression on proliferation, apoptosis, cell cycle, ERK1/2 and AKT phosphorylation, dabrafenib sensitivity, invasiveness and VEGF-A secretion were evaluated in the dabrafenib-resistant sublines using MTT assays, flow cytometry, immunoblotting, invasion assays in Boyden chambers and ELISA. ADAM9, PIK3R2, MMP7 and CXCR4 expression in the sensitive and dabrafenib-resistant cells was determined by immunoblotting. Small RNA interference was performed to investigate the consequence of VEGFA or ADAM9 silencing on proliferation, invasiveness or dabrafenib sensitivity of the resistant sublines. Long-term proliferation assays were carried out in dabrafenib-sensitive cells to assess the effects of enforced miR-126-3p expression or ADAM9 silencing on resistance development. VEGF-A serum levels in melanoma patients treated with BRAFi or BRAFi+MEKi were evaluated at baseline (T0), after two months of treatment (T2) and at progression (TP) by ELISA. Results miR-126-3p was significantly down-regulated in the dabrafenib-resistant sublines as compared with their parental counterparts. miR-126-3p replacement in the drug-resistant cells inhibited proliferation, cell cycle progression, phosphorylation of ERK1/2 and/or AKT, invasiveness, VEGF-A and ADAM9 expression, and increased dabrafenib sensitivity. VEGFA or ADAM9 silencing impaired proliferation and invasiveness of the drug-resistant sublines. ADAM9 knock-down in the resistant cells increased dabrafenib sensitivity, whereas miR-126-3p enforced expression or ADAM9 silencing in the drug-sensitive cells delayed the development of resistance. At T0 and T2, statistically significant differences were observed in VEGF-A serum levels between patients who responded to therapy and patients who did not. In responder patients, a significant increase of VEGF-A levels was observed at TP versus T2. Conclusions Strategies restoring miR-126-3p expression or targeting VEGF-A or ADAM9 could restrain growth and metastasis of dabrafenib-resistant melanomas and increase their drug sensitivity. Circulating VEGF-A is a promising biomarker for predicting patients’ response to BRAFi or BRAFi+MEKi and for monitoring the onset of resistance. Electronic supplementary material The online version of this article (10.1186/s13046-019-1238-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Simona Caporali
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167, Rome, Italy
| | - Adriana Amaro
- Molecular Pathology, IRCCS-Ospedale Policlinico San Martino, Genoa, Italy
| | - Lauretta Levati
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167, Rome, Italy
| | - Ester Alvino
- Institute of Translational Pharmacology, National Council of Research, Rome, Italy
| | - Pedro Miguel Lacal
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167, Rome, Italy
| | | | - Federica Ruffini
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167, Rome, Italy
| | - Laura Bonmassar
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167, Rome, Italy
| | | | - Nadia Felli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Carè
- Center of Gender Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Ulrich Pfeffer
- Molecular Pathology, IRCCS-Ospedale Policlinico San Martino, Genoa, Italy
| | - Stefania D'Atri
- Laboratory of Molecular Oncology, Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Via dei Monti di Creta 104, 00167, Rome, Italy.
| |
Collapse
|
20
|
Ou YC, Li JR, Wang JD, Chang CY, Wu CC, Chen WY, Kuan YH, Liao SL, Lu HC, Chen CJ. Fibronectin Promotes Cell Growth and Migration in Human Renal Cell Carcinoma Cells. Int J Mol Sci 2019; 20:2792. [PMID: 31181623 PMCID: PMC6600362 DOI: 10.3390/ijms20112792] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 01/22/2023] Open
Abstract
The prognostic and therapeutic values of fibronectin have been reported in patients with renal cell carcinoma (RCC). However, the underlying mechanisms of malignancy in RCC are not completely understood. We found that silencing of fibronectin expression attenuated human RCC 786-O and Caki-1 cell growth and migration. Silencing of potential fibronectin receptor integrin α5 and integrin β1 decreased 786-O cell ability in movement and chemotactic migration. Biochemical examination revealed a reduction of cyclin D1 and vimentin expression, transforming growth factor-β1 (TGF-β1) production, as well as Src and Smad phosphorylation in fibronectin-silenced 786-O and Caki-1 cells. Pharmacological inhibition of Src decreased 786-O cell growth and migration accompanied by a reduction of cyclin D1, fibronectin, vimentin, and TGF-β1 expression, as well as Src and Smad phosphorylation. In 786-O cells, higher activities in cell growth and migration than in Caki-1 cells were noted, along with elevated fibronectin and TGF-β1 expression. The additions of exogenous fibronectin and TGF-β1 promoted Caki-1 cell growth and migration, and increased cyclin D1, fibronectin, vimentin, and TGF-β1 expression, as well as Src and Smad phosphorylation. These findings highlight the role of fibronectin in RCC cell growth and migration involving Src and TGF-β1 signaling.
Collapse
Affiliation(s)
- Yen-Chuan Ou
- Department of Urology, Tungs' Taichung MetroHarbor Hospital, Taichung 435, Taiwan.
| | - Jian-Ri Li
- Division of Urology, Taichung Veterans General Hospital, Taichung 407, Taiwan.
| | - Jiaan-Der Wang
- Department of Children's Medical center, Taichung Veterans General Hospital, Taichung 407, Taiwan.
| | - Cheng-Yi Chang
- Department of Surgery, Feng Yuan Hospital, Taichung 420, Taiwan.
| | - Chih-Cheng Wu
- Department of Anesthesiology, Taichung Veterans General Hospital, Taichung 407, Taiwan.
- Department of Financial Engineering, Providence University, Taichung 433, Taiwan.
- Department of Data Science and Big Data Analytics, Providence University, Taichung 433, Taiwan.
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung 402, Taiwan.
| | - Yu-Hsiang Kuan
- Department of Pharmacology, Chung Shan Medical University, Taichung 402, Taiwan.
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan.
| | - Hsi-Chi Lu
- Food Science Department and Graduate Institute, Tunghai University, Taichung 407, Taiwan.
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 407, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
21
|
Tarhini AA, Frankel P, Ruel C, Ernstoff MS, Kuzel TM, Logan TF, Khushalani NI, Tawbi HA, Margolin KA, Awasthi S, Butterfield LH, McDermott D, Chen A, Lara PN, Kirkwood JM. NCI 8628: A randomized phase 2 study of ziv-aflibercept and high-dose interleukin 2 or high-dose interleukin 2 alone for inoperable stage III or IV melanoma. Cancer 2018; 124:4332-4341. [PMID: 30303516 PMCID: PMC6504933 DOI: 10.1002/cncr.31734] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 08/22/2016] [Accepted: 10/31/2016] [Indexed: 11/09/2022]
Abstract
BACKGROUND Interleukin 2 (IL-2) is a growth factor for T and natural killer cells, promotes proinflammatory cytokines, and can lead to durable responses in patients with melanoma. Vascular endothelial growth factor (VEGF) promotes angiogenesis and modulates host innate and adaptive immunity. High VEGF levels were found to be associated with nonresponse to IL-2. Ziv-aflibercept may deplete VEGF and thereby enhance antitumor T-cell responses, thus supporting a combination immunotherapeutic strategy with IL-2. METHODS NCI 8628 was a phase 2 trial of ziv-aflibercept and IL-2 (arm A) versus IL-2 alone (arm B) randomized at 2:1, respectively. Eligible patients had inoperable American Joint Committee on Cancer stage III or stage IV melanoma. The primary endpoint was progression-free survival (PFS). RESULTS A total of 89 patients were enrolled and 84 patients were treated. The median follow-up was 41.4 months. Among treated patients (55 patients in arm A and 29 patients in arm B), PFS was significantly improved in favor of arm A, with a median of 6.9 months (95% confidence interval [95% CI], 4.1-8.7 months) versus 2.3 months (95% CI, 1.6-3.5 months) (P<.001). No significant difference was noted with regard to overall survival, with a median of 26.9 months (95% CI, 14.4-63.6 months) for arm A and 24.2 months (95% CI, 11.3-36.4 months) for arm B. The response rate (according to Response Evaluation Criteria In Solid Tumors [RECIST]) was 22% in arm A (4 complete responses [CRs] and 8 partial responses [PRs]) and 17% in arm B (1 CR and 4 PRs). Stable disease or PR or CR was noted in 65% of patients in arm A and 48% of patients in arm B. The combination was found to be superior to monotherapy in patients with high and low levels of serum VEGF and VEGF receptor 2. Adverse events were consistent with the expected profiles of monotherapy with IL-2 and ziv-aflibercept. CONCLUSIONS Ziv-aflibercept and IL-2 were found to significantly improve PFS compared with IL-2 alone, thereby meeting the primary endpoint of the current study. These findings support further study of immunotherapeutic combination strategies involving VEGF inhibitors.
Collapse
Affiliation(s)
- Ahmad A. Tarhini
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Cleveland Clinic and Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Paul Frankel
- City of Hope National Medical Center, Duarte, CA, USA
| | | | | | | | | | | | - Hussein A. Tawbi
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Lisa H. Butterfield
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Departments of Medicine, Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Alice Chen
- Cancer Therapy Evaluation Program (CTEP), National Cancer Institute, Bethesda, MD, USA
| | - Primo N. Lara
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | | |
Collapse
|
22
|
Music M, Prassas I, Diamandis EP. Optimizing cancer immunotherapy: Is it time for personalized predictive biomarkers? Crit Rev Clin Lab Sci 2018; 55:466-479. [PMID: 30277835 DOI: 10.1080/10408363.2018.1499706] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cancer immunotherapy, a treatment that selectively augments a patient's anti-tumor immune response, is a breakthrough advancement in personalized medicine. A subset of cancer patients undergoing immunotherapy have displayed robust and long-lasting therapeutic responses. Currently, the spotlight is on the use of blocking antibodies against the T-cell checkpoint molecules, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programed cell death-1 (PD-1)/programed death-ligand 1 (PD-L1), which have been effectively used to combat many cancers types. Despite the overall enthusiasm, immune checkpoint blockade inhibitors suffer from significant limitations such as high cost, serious toxicity in a substantial proportion of patients, and a response rate as low as 10%-40% in some clinical trials. Consequently, there is an urgent and unmet medical need for companion biomarkers that could both predict the response of individual patients to these therapies, and provide the means for precise monitoring of their therapeutic outcome. In this era of precision medicine, predictive biomarkers are a hot commodity because they can effectively separate responders from non-responders, and spare non-responders from serious therapy-related toxicity. Emerging predictive biomarkers for immune checkpoint blockade are: PD-L1 expression, increased amounts of tumor-infiltrating lymphocytes, increased mutational load and mismatch repair deficiency. Other well-studied biomarkers include inflammatory infiltrate, absolute lymphocyte count and lactate dehydrogenase levels. We review recent progress on predictive cancer biomarkers in immunotherapy, with a special emphasis on serum autoantibodies that have the potential to be personalized for optimal clinical outcomes.
Collapse
Affiliation(s)
- Milena Music
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada
| | - Ioannis Prassas
- b Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , Canada
| | - Eleftherios P Diamandis
- a Department of Laboratory Medicine and Pathobiology , University of Toronto , Toronto , Canada.,b Department of Pathology and Laboratory Medicine , Mount Sinai Hospital , Toronto , Canada.,c Department of Clinical Biochemistry , University Health Network , Toronto , Canada.,d Lunenfeld-Tanenbaum Research Institute , Mount Sinai Hospital , Toronto , Canada
| |
Collapse
|
23
|
Duskova K, Vesely S, DO Carmo Silva J, Cernei N, Zitka O, Heger Z, Adam V, Havlova K, Babjuk M. Differences in Urinary Amino Acid Patterns in Individuals with Different Types of Urological Tumor Urinary Amino Acid Patterns as Markers of Urological Tumors. ACTA ACUST UNITED AC 2018; 32:425-429. [PMID: 29475932 DOI: 10.21873/invivo.11257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 01/09/2018] [Accepted: 01/10/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Insufficient specificity and invasiveness of currently used diagnostic methods raises the need for new markers of urological tumors. The aim of this study was to find a link between the urinary excretion of amino acids and the presence of urological tumors. MATERIALS AND METHODS Using ion-exchange chromatography, we tested urine samples of patients with prostate cancer (n=30), urinary bladder cancer (n=28), renal cell carcinoma (n=16) and healthy volunteers (control group; n=21). RESULTS In each category, we found a group of amino acids which differed in concentration compared to the control group. These differences were most significant in sarcosine in patients with prostate cancer; leucine, phenylalanine and arginine in those with bladder cancer; and sarcosine, glutamic acid, glycine, tyrosine and arginine in the those with renal cell carcinoma. CONCLUSION Results of our research imply a possible connection between the occurrence of specific types of amino acids in the urine and the presence of urological tumors.
Collapse
Affiliation(s)
- Katerina Duskova
- Department of Urology, Second Faculty of Medicine, Charles University, and University Hospital Motol, Prague, Czech Republic
| | - Stepan Vesely
- Department of Urology, Second Faculty of Medicine, Charles University, and University Hospital Motol, Prague, Czech Republic
| | - Joana DO Carmo Silva
- Department of Urology, Second Faculty of Medicine, Charles University, and University Hospital Motol, Prague, Czech Republic
| | - Natalia Cernei
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic.,Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic
| | - Ondrej Zitka
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic.,Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic
| | - Zbynek Heger
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic.,Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic
| | - Vojtech Adam
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic.,Department of Chemistry and Biochemistry, Mendel University in Brno, Brno, Czech Republic
| | - Klara Havlova
- Department of Urology, Second Faculty of Medicine, Charles University, and University Hospital Motol, Prague, Czech Republic
| | - Marek Babjuk
- Department of Urology, Second Faculty of Medicine, Charles University, and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
24
|
Immune Profiling of Cancer Patients Treated with Immunotherapy: Advances and Challenges. Biomedicines 2018; 6:biomedicines6030076. [PMID: 30004433 PMCID: PMC6163220 DOI: 10.3390/biomedicines6030076] [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: 05/28/2018] [Revised: 06/23/2018] [Accepted: 06/25/2018] [Indexed: 12/16/2022] Open
Abstract
The recent advances in immunotherapy and the availability of novel drugs to target the tumor microenvironment have dramatically changed the paradigm of cancer treatment. Nevertheless, a significant proportion of cancer patients are unresponsive or develop resistance to these treatments. With the aim to increase the clinical efficacy of immunotherapy, combinations of agents and standard therapies with complementary actions have been developed mostly on an empirical base, since their mechanisms of actions are not yet fully dissected. The characterization of immune responsiveness and its monitoring along with the treatment of cancer patients with immunotherapy can provide insights into the mechanisms of action of these therapeutic regimens and contribute to the optimization of patients’ stratification and of combination strategies and to the prediction of treatment-related toxicities. Thus far, none of the immunomonitoring strategies has been validated for routine clinical practice. Moreover, it is becoming clear that the genomic and molecular make-up of tumors and of the infiltrating immune system represent important determinants of the clinical responses to immunotherapy. This review provides an overview of different approaches for the immune profiling of cancer patients and discusses their advantages and limitations. Recent advances in genomic-based assays and in the identification of host genomic relationships with immune responses represent promising approaches to identify molecular determinants and biomarkers to improve the clinical efficacy of cancer immunotherapy.
Collapse
|
25
|
Mytsyk Y, Dosenko V, Skrzypczyk MA, Borys Y, Diychuk Y, Kucher A, Kowalskyy V, Pasichnyk S, Mytsyk O, Manyuk L. Potential clinical applications of microRNAs as biomarkers for renal cell carcinoma. Cent European J Urol 2018; 71:295-303. [PMID: 30386650 PMCID: PMC6202627 DOI: 10.5173/ceju.2018.1618] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/26/2018] [Accepted: 06/25/2018] [Indexed: 12/31/2022] Open
Abstract
Introduction Renal cell carcinoma (RCC) accounts for 3% of adult malignancies and more than 90% of kidney neoplasms. High rates of undiagnostic percutaneous kidney biopsies and difficulties in reliable pre-operative differentiation between malignant and benign renal tumors using contemporary imaging techniques result in large numbers of redundant surgeries. Absence of specific biomarkers for early detection and monitoring complicates on-time diagnosis of the disease and relapse. For the patients followed up after having a nephrectomy, a noninvasive and sensitive biomarker enabling early detection of disease relapse would be extremely useful. Material and methods The study is a review of recent knowledge regarding potential clinical applications of microRNAs (miRNAs) as biomarkers of RCC. Results MicroRNAs are essential regulators of various processes such as cell proliferation, differentiation, development and death; they have been implicated in diverse biological and pathological processes in RCC. There is a class of miRNAs that promote RCC development (oncomirs) and a class of miRNAs that negatively regulate oncogenes, suppress tumor growth and invasion, and thus could be considered treatment agents (anti-oncomirs). Separate miRNAs and specific miRNAs expression profiles have been identified, enabling early detection of the disease, prediction of response to systemic therapy, or prognostication of biological behavior of the disease. Conclusions The miRNA network analysis and gene profiling may help to identify the most sensible molecular signatures of RCC that can be used for diagnostic purposes, as well as poor prognosis signatures and poor therapeutic response signatures in patients who undergo systemic therapy.
Collapse
Affiliation(s)
- Yulian Mytsyk
- Department of Urology, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Victor Dosenko
- General and Molecular Pathophysiology Department, Bogomoletz Institute of Physiology of National Academy of Sciences of Ukraine, Kiev, Ukraine
| | | | - Yuriy Borys
- Department of Urology, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Yuriy Diychuk
- Department of Urology, Lviv State Regional Oncology Treatment and Diagnostic Center, Lviv, Ukraine
| | - Askold Kucher
- Department of Urology, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Vasyl Kowalskyy
- Department of Urology, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Serhyi Pasichnyk
- Department of Urology, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Oleh Mytsyk
- Lviv Regional Bureau of Forensic Services, Lviv, Ukraine
| | - Lubov Manyuk
- Department of Foreign Languages, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| |
Collapse
|
26
|
Abstract
Melanoma represents the most aggressive and the deadliest form of skin cancer. Current therapeutic approaches include surgical resection, chemotherapy, photodynamic therapy, immunotherapy, biochemotherapy, and targeted therapy. The therapeutic strategy can include single agents or combined therapies, depending on the patient’s health, stage, and location of the tumor. The efficiency of these treatments can be decreased due to the development of diverse resistance mechanisms. New therapeutic targets have emerged from studies of the genetic profile of melanocytes and from the identification of molecular factors involved in the pathogenesis of the malignant transformation. In this review, we aim to survey therapies approved and under evaluation for melanoma treatment and relevant research on the molecular mechanisms underlying melanomagenesis.
Collapse
Affiliation(s)
- Beatriz Domingues
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Faculty of Sciences, University of Porto, Porto, Portugal
| | - José Manuel Lopes
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Department of Pathology, Hospital S João, Porto, Portugal.,Department of Pathology, Medical Faculty, University of Porto, Porto, Portugal
| | - Paula Soares
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Department of Pathology, Medical Faculty, University of Porto, Porto, Portugal
| | - Helena Pópulo
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Porto, Portugal.,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
27
|
Hoeres T, Smetak M, Pretscher D, Wilhelm M. Improving the Efficiency of Vγ9Vδ2 T-Cell Immunotherapy in Cancer. Front Immunol 2018; 9:800. [PMID: 29725332 PMCID: PMC5916964 DOI: 10.3389/fimmu.2018.00800] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/03/2018] [Indexed: 12/28/2022] Open
Abstract
Increasing immunological knowledge and advances in techniques lay the ground for more efficient and broader application of immunotherapies. gamma delta (γδ) T-cells possess multiple favorable anti-tumor characteristics, making them promising candidates to be used in cellular and combination therapies of cancer. They recognize malignant cells, infiltrate tumors, and depict strong cytotoxic and pro-inflammatory activity. Here, we focus on human Vγ9Vδ2 T-cells, the most abundant γδ T-cell subpopulation in the blood, which are able to inhibit cancer progression in various models in vitro and in vivo. For therapeutic use they can be cultured and manipulated ex vivo and in the following adoptively transferred to patients, as well as directly stimulated to propagate in vivo. In clinical studies, Vγ9Vδ2 T-cells repeatedly demonstrated a low toxicity profile but hitherto only the modest therapeutic efficacy. This review provides a comprehensive summary of established and newer strategies for the enhancement of Vγ9Vδ2 T-cell anti-tumor functions. We discuss data of studies exploring methods for the sensitization of malignant cells, the improvement of recognition mechanisms and cytotoxic activity of Vγ9Vδ2 T-cells. Main aspects are the tumor cell metabolism, antibody-dependent cell-mediated cytotoxicity, antibody constructs, as well as activating and inhibitory receptors like NKG2D and immune checkpoint molecules. Several concepts show promising results in vitro, now awaiting translation to in vivo models and clinical studies. Given the array of research and encouraging findings in this area, this review aims at optimizing future investigations, specifically targeting the unanswered questions.
Collapse
Affiliation(s)
- Timm Hoeres
- Department of Hematology and Medical Oncology, Paracelsus Medical University, Nuremberg, Germany
| | - Manfred Smetak
- Department of Hematology and Medical Oncology, Paracelsus Medical University, Nuremberg, Germany
| | - Dominik Pretscher
- Department of Hematology and Medical Oncology, Paracelsus Medical University, Nuremberg, Germany
| | - Martin Wilhelm
- Department of Hematology and Medical Oncology, Paracelsus Medical University, Nuremberg, Germany
| |
Collapse
|
28
|
MicroRNA-15a expression measured in urine samples as a potential biomarker of renal cell carcinoma. Int Urol Nephrol 2018; 50:851-859. [DOI: 10.1007/s11255-018-1841-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/07/2018] [Indexed: 01/18/2023]
|
29
|
Hoeres T, Wilhelm M, Smetak M, Holzmann E, Schulze-Tanzil G, Birkmann J. Immune cells regulate VEGF signalling via release of VEGF and antagonistic soluble VEGF receptor-1. Clin Exp Immunol 2018; 192:54-67. [PMID: 29235095 DOI: 10.1111/cei.13090] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/03/2017] [Accepted: 11/28/2017] [Indexed: 12/12/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is an important regulator of physiological and pathological angiogenesis. Besides malignant and stromal cells, local immune cells shape VEGF signalling in the tumour microenvironment. Aminobisphosphonates such as zoledronic acid (Zol) are drugs known to inhibit osteoclast activity and bone resorption, but also have immunomodulatory and anti-tumour effects. These properties have been linked previously to the down-regulation of VEGF and interference with tumour neo-angiogenesis. It was therefore surprising to find that treatment with Zol in combination with low-dose interleukin (IL)-2 increased serum VEGF levels in cancer patients. In this study we aimed to characterize the effect of Zol and IL-2 on VEGF signalling of blood-derived immune cells in vitro. Upon stimulation with IL-2, T cells and natural killer (NK) cells increase production of VEGF consecutively to the release of proinflammatory interferon (IFN)-γ, and Zol accelerates this response specifically in γδ T cells. VEGF can, in turn, be antagonized by soluble VEGF receptor (sVEGFR)-1, which is released depending on stimulatory conditions and the presence of monocytes. Additionally, malignant cells represented by leukaemia and lymphoma cell lines produce VEGF and some release sVEGFR-1 simultaneously. Our findings indicate a mechanism by which the VEGF and the sVEGFR-1 production by immune cells regulates local VEGF signalling. Therefore, immunotherapeutic interventions may enable both pro- as well as anti-tumour effects via immune cell-mediated alterations of VEGF homeostasis.
Collapse
Affiliation(s)
- T Hoeres
- Department of Hematology and Medical Oncology, Paracelsus Medical University, Nuremberg, Germany
| | - M Wilhelm
- Department of Hematology and Medical Oncology, Paracelsus Medical University, Nuremberg, Germany
| | - M Smetak
- Department of Hematology and Medical Oncology, Paracelsus Medical University, Nuremberg, Germany
| | - E Holzmann
- Department of Hematology and Medical Oncology, Paracelsus Medical University, Nuremberg, Germany
| | - G Schulze-Tanzil
- Institute of Anatomy, Paracelsus Medical University, Nuremberg, Germany
| | - J Birkmann
- Department of Hematology and Medical Oncology, Paracelsus Medical University, Nuremberg, Germany
| |
Collapse
|
30
|
Abstract
The relatively high DNA mutational burden in melanoma allows for the creation of potentially "foreign," immune-stimulating neoantigens, and leads to its exceptional immunogenicity. Brisk tumor-infiltrating lymphocytes, a marker of immune editing, confer improved overall survival in melanoma, possibly due to reduced sentinel lymph node spread. Meanwhile, T-cell-stimulating drugs, so-called T-cell checkpoint inhibitors, which reverse peripheral tolerance-dependent tumor escape, have demonstrated unparalleled clinical success in metastatic melanoma. Markers to predict response to immunotherapy are currently imperfect, and the subject of intense research, which will guide the future of ancillary pathologic testing in this setting.
Collapse
Affiliation(s)
- Jennifer S Ko
- Department of Anatomic Pathology, Cleveland Clinic, 9500 Euclid Avenue, L2-150, Cleveland, OH 44195, USA.
| |
Collapse
|
31
|
Marabondo S, Kaufman HL. High-dose interleukin-2 (IL-2) for the treatment of melanoma: safety considerations and future directions. Expert Opin Drug Saf 2017; 16:1347-1357. [PMID: 28929820 DOI: 10.1080/14740338.2017.1382472] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION In 1998, high-dose interleukin-2 (IL-2) was the first immunotherapy approved for the treatment of metastatic melanoma based on durable objective responses documented in a subset of patients but widespread utilization was limited by significant toxicity. Advances in targeted therapy and the emergence of T cell checkpoint inhibitors, which can generally be given in the ambulatory setting, have further limited consideration of IL-2 for melanoma patients and the role of IL-2 in the current landscape of melanoma treatment is uncertain. Areas covered: In this review, we will describe advances in clinical diagnostic and management strategies that have improved the therapeutic window for IL-2 therapy in patients with melanoma. Further, we will describe the potential for using IL-2 in patients whose disease has progressed after other interventions or as part of combination immunotherapy approaches that are now in clinical development. We will also review the common toxicities of IL-2 therapy and their current management will be discussed. Expert opinion: High-dose IL-2 remains an important option for patients with melanoma and has an improved therapeutic window in the contemporary era. The reasons why IL-2 is not utilized more frequently and measures for enhancing its use will be detailed.
Collapse
Affiliation(s)
- Stephen Marabondo
- a Departments of Surgery and Medicine, Rutgers Robert Wood Johnson Medical School , Rutgers University , New Brunswick , NJ , USA
| | - Howard L Kaufman
- a Departments of Surgery and Medicine, Rutgers Robert Wood Johnson Medical School , Rutgers University , New Brunswick , NJ , USA
| |
Collapse
|
32
|
Anselmo Da Costa I, Rausch S, Kruck S, Todenhöfer T, Stenzl A, Bedke J. Immunotherapeutic strategies for the treatment of renal cell carcinoma: Where will we go? Expert Rev Anticancer Ther 2017; 17:357-368. [PMID: 28162024 DOI: 10.1080/14737140.2017.1292138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Historically, renal cell carcinoma (RCC) is considered a chemotherapy-resistant tumor. The cornerstone of systemic therapy included mammalian target of rapamycin (mTOR) inhibitors, endothelial growth factor receptor (VEGFR) and tyrosine kinase inhibitors (TKIs). Currently, a new era is enteres with promising immunotherapeutic treatments, which are becoming commercially available. Areas covered: We provide a comprehensive review using PubMed and ClinicalTrials.gov about the following immunotherapies in RCC: i) vaccine therapy, ii) adoptive T Cell Transfer and CAR T cells, iii) nonspecific immunotherapy - IL-2 (new formulations), iv) Checkpoint inhibitors, v) other checkpoint-molecules. We will also discuss their mechanism of action and toxicity, the importance of developing new patient selection algorithms (immunoprofiling, guidelines updates) and new biomarkers such as PD-1 expression. Expert commentary: Immunotherapy shows promise, and the current tools used in clinical practice, including guidelines, staging-classification and algorithms should be revised and adapted to the new immunotherapeutic drugs. Although immunotherapy in RCC show promising results, more research is needed in parallel to discover biomarkers that enable the prediction of a treatment response and therefore lead to better patient selection.
Collapse
Affiliation(s)
| | - Steffen Rausch
- a Department of Urology , Eberhard Karls University , Tuebingen , Germany
| | - Stephan Kruck
- a Department of Urology , Eberhard Karls University , Tuebingen , Germany
| | - Tilman Todenhöfer
- a Department of Urology , Eberhard Karls University , Tuebingen , Germany
| | - Arnulf Stenzl
- a Department of Urology , Eberhard Karls University , Tuebingen , Germany
| | - Jens Bedke
- a Department of Urology , Eberhard Karls University , Tuebingen , Germany
| |
Collapse
|
33
|
Spencer KR, Wang J, Silk AW, Ganesan S, Kaufman HL, Mehnert JM. Biomarkers for Immunotherapy: Current Developments and Challenges. Am Soc Clin Oncol Educ Book 2017; 35:e493-503. [PMID: 27249758 DOI: 10.1200/edbk_160766] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Immunotherapy has revolutionized cancer therapy and has been named the cancer advance of the year for 2016. Checkpoint inhibitors have demonstrated unprecedented rates of durable responses in some of the most difficult-to-treat cancers; however, many treated patients do not respond, and the potential for serious side effects exists. There is a growing need to identify biomarkers that will improve the selection of patients who will best respond to therapy, further elucidate drug mechanisms of action, and help tailor therapy regimens. Biomarkers are being explored at the soluble, cellular, and genomic levels, and examples in immunotherapy include serum proteins, tumor-specific receptor expression patterns, factors in the tumor microenvironment, circulating immune and tumor cells, and host genomic factors. The search for reliable biomarkers is limited by our incomplete understanding of how immunotherapies modify the already complex immune response to cancer, as well as the contribution of immuno-editing to a dynamic and inducible tumor microenvironment and immune milieu. Furthermore, there has been little extension of any candidate assay into large, prospective studies, and the lack of standardization in measurement and interpretation restricts their validity. Both tumor-infiltrating lymphocytes and PD-L1 expression within the tumor microenvironment have been recognized as having both prognostic and predictive value for patients treated with immunotherapy. Alternately, a larger panel of gene signatures, chemokines, and other factors that correlate with response has been proposed. In this article, we will explore the status of current biomarker candidates.
Collapse
Affiliation(s)
| | - Jianfeng Wang
- From the Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Ann W Silk
- From the Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Shridar Ganesan
- From the Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Howard L Kaufman
- From the Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Janice M Mehnert
- From the Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| |
Collapse
|
34
|
Zloza A, Dharmadhikari ND, Huelsmann EJ, Broucek JR, Hughes T, Kohlhapp FJ, Kaufman HL. Low-dose interleukin-2 impairs host anti-tumor immunity and inhibits therapeutic responses in a mouse model of melanoma. Cancer Immunol Immunother 2017; 66:9-16. [PMID: 27757560 PMCID: PMC11028934 DOI: 10.1007/s00262-016-1916-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 10/12/2016] [Indexed: 12/25/2022]
Abstract
Recombinant interleukin-2 (rIL-2) is associated with objective responses in 15-20 % of patients with metastatic melanoma and renal cell carcinoma. More recently, rIL-2 has also demonstrated improved clinical activity in patients with melanoma. Given the toxicity of high-dose rIL-2 and the availability of many new immunotherapy agents, it has been suggested that lower doses of rIL-2 may be preferred for combination clinical studies. In order to determine the impact of low doses of rIL-2 on anti-tumor immunity and therapeutic effectiveness, we challenged C57BL/6 mice with poorly immunogenic B16-F10 melanoma and treated them with varying doses of rIL-2 (range 103-105 IU). Tumor growth at day 14 was significantly reduced when rIL-2 was administered at 10,000 (P < 0.02) and 100,000 (P < 0.02) IU doses, but tumor growth was significantly increased when mice were treated at 1000 IU rIL-2 (P < 0.02), as compared to placebo treatment. While the proportions of CD8+ and CD4+ T cells in the tumor were similar at all doses tested, the proportion of NK cells was decreased and the proportion of Tregs was increased in tumors exposed to low-dose rIL-2. The ratio of gp100-specific CD8+ to CD4+ regulatory T cells was increased in tumors treated at 10,000 and 100,000 IU of rIL-2 but was decreased at the 1000 IU dose compared to placebo-treated mice. These findings suggest that low-dose rIL-2 may impair host anti-tumor immunity and promote tumor growth. Early-phase adjuvant and combination clinical studies should include patient cohorts with higher doses of rIL-2.
Collapse
Affiliation(s)
- Andrew Zloza
- Division of Surgical Oncology Research, Section of Surgical Oncology, Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ, 08903, USA
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL, USA
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Neal D Dharmadhikari
- Division of Surgical Oncology Research, Section of Surgical Oncology, Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ, 08903, USA
| | - Erica J Huelsmann
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, IL, USA
| | - Joseph R Broucek
- Department of General Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Tasha Hughes
- Department of General Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Frederick J Kohlhapp
- Division of Surgical Oncology Research, Section of Surgical Oncology, Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ, 08903, USA
| | - Howard L Kaufman
- Division of Surgical Oncology Research, Section of Surgical Oncology, Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ, 08903, USA.
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
35
|
Harrington KJ, Puzanov I, Hecht JR, Hodi FS, Szabo Z, Murugappan S, Kaufman HL. Clinical development of talimogene laherparepvec (T-VEC): a modified herpes simplex virus type-1-derived oncolytic immunotherapy. Expert Rev Anticancer Ther 2016; 15:1389-403. [PMID: 26558498 DOI: 10.1586/14737140.2015.1115725] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tumor immunotherapy is emerging as a promising new treatment option for patients with cancer. T-VEC is an intralesional oncolytic virus therapy based on a modified herpes simplex virus type-1. T-VEC selectively targets tumor cells, causing regression in injected lesions and inducing immunologic responses that mediate regression at uninjected/distant sites. In a randomized phase III trial, T-VEC met its primary endpoint of improving the durable response rate vs granulocyte-macrophage colony-stimulating factor in patients with unresectable melanoma. Responses were observed in injected and uninjected regional and visceral lesions. Exploratory analyses suggested survival differences in favor of T-VEC in patients with untreated or stage IIIB/IIIC/IVM1a disease. T-VEC was generally well tolerated, the most common adverse events being flu-like symptoms. Here, we overview recent advances in cancer immunotherapy, focusing on the clinical development of T-VEC, from first-in-human studies and studies in other cancer types, to ongoing combination trials with checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Igor Puzanov
- a Division of Hematology-Oncology, Vanderbilt University Medical Center , Nashville , TN , USA
| | - J Randolph Hecht
- b David Geffen School of Medicine , UCLA , Los Angeles , CA , USA
| | - F Stephen Hodi
- c Melanoma Center and the Center for Immuno-Oncology , Dana-Farber Cancer Institute , Boston , MA , USA
| | - Zsolt Szabo
- d Department of Oncology , Amgen (Europe) GmbH , Zug , Switzerland
| | - Swami Murugappan
- e Department of Oncology , Amgen Inc ., Thousand Oaks , CA , USA
| | - Howard L Kaufman
- f Division of Surgical Oncology , Rutgers Cancer Institute of New Jersey , New Brunswick , NJ , USA
| |
Collapse
|
36
|
Sridharan V, Margalit DN, Lynch SA, Severgnini M, Hodi FS, Haddad RI, Tishler RB, Schoenfeld JD. Effects of definitive chemoradiation on circulating immunologic angiogenic cytokines in head and neck cancer patients. J Immunother Cancer 2016; 4:32. [PMID: 27330805 PMCID: PMC4915184 DOI: 10.1186/s40425-016-0138-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 05/17/2016] [Indexed: 12/28/2022] Open
Abstract
Background Preclinical studies suggest a synergistic effect between radiation, immunotherapy and anti-angiogenic therapy, although the mechanisms are unclear. Angiogenic cytokines are known to affect the immune system, and their levels may be associated with response to immunotherapy. Here, we assess changes in circulating VEGF, as well as angiogenic cytokines angiopoietin-1 and -2 (Ang1, Ang2), and placental growth factor (PLGF) that occur during definitive chemo-radiotherapy in HNSCC patients. Methods We prospectively collected blood samples from patients receiving definitive radiation with or without chemotherapy. Serum Ang1, Ang2, VEGF, and PLGF were measured via cytokine assays. Results The majority of patients had advanced stage, node positive HPV-associated oropharyngeal cancer, and received radiation to a median dose of 70 Gy with concurrent cisplatin. Over the course of treatment, serum VEGF and Ang1 levels decreased in 20/24 (84 %, p < 0.0001) and 21/24 (88 %, p < 0.0001) patients, respectively, and Ang2 and PLGF levels increased in 20/24 (83 %, p < 0.0001) patients. Conclusions We find significant changes in angiogenic cytokines in the majority of HNSCC patients over the course of chemoradiation. Decreases in VEGF caused by radiation may represent one mechanism of potential synergy with immunotherapy. Increases in Ang2 and PLGF are interesting given their link to tumor associated angiogenesis and poor prognosis. Additional studies are needed to explore synergies between anti-angiogenic treatments, immunotherapy, and chemoradiation in HNSCC. Electronic supplementary material The online version of this article (doi:10.1186/s40425-016-0138-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vishwajith Sridharan
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Center, 450 Brookline Ave, DA L2-57, Boston, MA 02114 USA ; Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA USA
| | - Danielle N Margalit
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Center, 450 Brookline Ave, DA L2-57, Boston, MA 02114 USA
| | - Stephanie A Lynch
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Center, 450 Brookline Ave, DA L2-57, Boston, MA 02114 USA
| | - Mariano Severgnini
- Center for Immuno-oncology, Dana-Farber Cancer Institute, Boston, MA USA
| | - F Stephen Hodi
- Center for Immuno-oncology, Dana-Farber Cancer Institute, Boston, MA USA ; Department of Medical Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Center, Boston, MA USA
| | - Robert I Haddad
- Department of Medical Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Center, Boston, MA USA
| | - Roy B Tishler
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Center, 450 Brookline Ave, DA L2-57, Boston, MA 02114 USA
| | - Jonathan D Schoenfeld
- Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Center, 450 Brookline Ave, DA L2-57, Boston, MA 02114 USA
| |
Collapse
|
37
|
Buchbinder EI, Flaherty KT. Biomarkers in Melanoma: Lessons from Translational Medicine. Trends Cancer 2016; 2:305-312. [PMID: 28741528 DOI: 10.1016/j.trecan.2016.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 05/10/2016] [Accepted: 05/11/2016] [Indexed: 12/19/2022]
Abstract
The treatment landscape for advanced melanoma has been rapidly evolving. As new therapies become available, there is a need for better biomarkers to detect disease, guide patient selection, and monitor for response. The use of tumor genetics has been able to predict responses to targeted therapy in melanoma. However, the role of biomarkers in melanoma detection, monitoring, and immunotherapy has been less successful and is still being defined. Translational studies in many areas of melanoma are being performed to identify biomarkers and validate their clinical role. In this review, we examine the status of biomarkers in melanoma and areas of future development.
Collapse
Affiliation(s)
| | - Keith T Flaherty
- Department of Hematology/Oncology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
38
|
Kim DW, Anderson J, Patel SP. Immunotherapy for uveal melanoma. Melanoma Manag 2016; 3:125-135. [PMID: 30190881 DOI: 10.2217/mmt-2015-0006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 04/13/2016] [Indexed: 11/21/2022] Open
Abstract
Uveal melanoma (UM) is a rare cancer with a high mortality rate. In comparison to cutaneous melanoma, UM has unique immunological features. Arising in the immune suppressive environment of the eye, it maintains immune resistance once metastatic. This is considered a major obstacle for successful immunotherapy in UM. However, a growing body of evidence suggests strategies that may abrogate resistance and enhance antitumor immunity in UM. Recently, three new immune agents have been approved for melanoma. While these drugs demonstrate durable clinical responses with long-term remissions in metastatic cutaneous melanoma, only limited data exist in metastatic UM. In this review, immunological aspects of UM and data from clinical studies of immunotherapeutic agents and regimens for UM will be discussed.
Collapse
Affiliation(s)
- Dae Won Kim
- Moffitt Cancer Center, Tampa, FL 33612, USA.,Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jaime Anderson
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd Unit 0430, Houston, TX 77030, USA.,Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd Unit 0430, Houston, TX 77030, USA
| | - Sapna P Patel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd Unit 0430, Houston, TX 77030, USA.,Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd Unit 0430, Houston, TX 77030, USA
| |
Collapse
|
39
|
Quiroga Matamoros W, Fernandez F, Citarella Otero D, Rangel J, Estrada Guerrero A, Patiño ID. Guía de manejo del carcinoma de células renales. Rev Urol 2016. [DOI: 10.1016/j.uroco.2016.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
40
|
Slovin SF. Biomarkers for immunotherapy in genitourinary malignancies. Urol Oncol 2016; 34:205-13. [PMID: 25791754 PMCID: PMC8675216 DOI: 10.1016/j.urolonc.2015.02.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 02/08/2015] [Accepted: 02/11/2015] [Indexed: 12/23/2022]
Abstract
Immunotherapy for genitourinary malignancies such as prostate, renal, and bladder cancers has experienced a resurgence since the development of 3 novel strategies: the autologous cellular product therapy, Sipuleucel-T for prostate cancer, the checkpoint inhibitors, anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4), anti-programmed cell death ligand 1 (anti-PD1), and anti-programmed cell death ligand 1), respectively. These agents have led to strikingly durable responses in several of these solid tumors, but their efficacy has been inconsistent. Why all solid tumors are not equal in their response to these therapies is unclear. More importantly, changes in humoral or cellular responses which may reflect changes in a tumor's biology have been limited due to differences in immune monitoring and lack of consistency in established reliable immunologic endpoints. How to design immunologic end points that reflect a meaningful effect on the cancer remains a challenge for clinical trial development. The issues faced by clinical investigators and the current state of immune monitoring are discussed.
Collapse
Affiliation(s)
- Susan F Slovin
- Genitourinary Oncology Service, Sidney Kimmel Center for Prostate and Urologic Cancers, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY.
| |
Collapse
|
41
|
Abstract
In recent years, the introduction and Federal Drug Administration approval of immune checkpoint inhibitor antibodies has dramatically improved the clinical outcomes for patients with advanced melanoma. These antagonist monoclonal antibodies are capable of unleashing dormant or exhausted antitumor immunity, which has led to durable complete and partial responses in a large number of patients. Ipilimumab targets the cytotoxic T lymphocyte-associated protein 4 (CTLA-4) receptor. Nivolumab and pembrolizumab target programmed cell death protein 1 (PD-1) receptors and have proven to be superior to ipilimumab alone. The combination of ipilimumab and nivolumab has yielded higher response rates, greater tumor shrinkage, and longer progression-free survival than either monotherapy alone. As other promising immunotherapies for melanoma proceed through clinical trials, future goals include defining the role of immune checkpoint inhibitors as adjuvant therapy, identifying optimal combination strategies, and developing reliable predictive biomarkers to guide treatment selection for individual patients.
Collapse
Affiliation(s)
- Jason M Redman
- Georgetown Lombardi Comprehensive Cancer Center 3970 Reservoir Road, NW Research Building, Room E501, 20007, Washington DC, USA. .,Department of Medicine, Georgetown University Medical Center, Washington DC, USA.
| | - Geoffrey T Gibney
- Georgetown Lombardi Comprehensive Cancer Center 3970 Reservoir Road, NW Research Building, Room E501, 20007, Washington DC, USA. .,Department of Medicine, Georgetown University Medical Center, Washington DC, USA.
| | - Michael B Atkins
- Georgetown Lombardi Comprehensive Cancer Center 3970 Reservoir Road, NW Research Building, Room E501, 20007, Washington DC, USA. .,Department of Medicine, Georgetown University Medical Center, Washington DC, USA.
| |
Collapse
|
42
|
Tumour-infiltrating lymphocytes in melanoma prognosis and cancer immunotherapy. Pathology 2016; 48:177-87. [PMID: 27020390 DOI: 10.1016/j.pathol.2015.12.006] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 11/16/2015] [Indexed: 12/26/2022]
Abstract
The field of systemic cancer therapy for metastatic disease has entered an exciting era with the advent of novel immunomodulatory strategies targeting immune checkpoints. At the heart of these promising efforts are the tumour-infiltrating lymphocytes (TILs). As the reports demonstrating efficacy of modulating TIL effector function in patients with advanced stage cancer continue to accrue, it has become essential to better understand TIL immunobiology in order to further improve clinical outcome. In addition to providing an overview of the current immunotherapies available for metastatic melanoma, this review will briefly introduce the history and classification of TILs. Moreover, we will dissect the multifaceted roles of TILs in tumour-specific immunity and melanoma immune escape. The significance of TILs in melanoma prognosis and cancer immunotherapy will also be discussed, with a particular focus on their potential utility as biomarkers of patient response. The goal of personalised medicine appears to be in realistic sight, as new immunomodulatory techniques and technological innovations continue to advance the field of cancer immunotherapy. In light of recent studies highlighting the possible utility of TILs in determining therapeutic outcome, further characterisation of TIL phenotype and function has the potential to help translate individualised care into current medical practice.
Collapse
|
43
|
Validation of the diagnostic and prognostic relevance of serum MMP-7 levels in renal cell cancer by using a novel automated fluorescent immunoassay method. Int Urol Nephrol 2016; 48:355-61. [DOI: 10.1007/s11255-015-1185-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/07/2015] [Indexed: 10/22/2022]
|
44
|
Activation and propagation of tumor-infiltrating lymphocytes on clinical-grade designer artificial antigen-presenting cells for adoptive immunotherapy of melanoma. J Immunother 2015; 37:448-60. [PMID: 25304728 DOI: 10.1097/cji.0000000000000056] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Adoptive cell therapy with autologous tumor-infiltrating lymphocytes (TIL) is a therapy for metastatic melanoma with response rates of up to 50%. However, the generation of the TIL transfer product is challenging, requiring pooled allogeneic normal donor peripheral blood mononuclear cells (PBMC) used in vitro as "feeders" to support a rapid-expansion protocol. Here, we optimized a platform to propagate TIL to a clinical scale using K562 cells genetically modified to express costimulatory molecules such as CD86, CD137-ligand, and membrane-bound IL-15 to function as artificial antigen-presenting cells (aAPC) as an alternative to using PBMC feeders. EXPERIMENTAL DESIGN We used aAPC or γ-irradiated PBMC feeders to propagate TIL and measured rates of expansion. The activation and differentiation state was evaluated by flow cytometry and differential gene expression analyses. Clonal diversity was assessed on the basis of the pattern of T-cell receptor usage. T-cell effector function was measured by evaluation of cytotoxic granule content and killing of target cells. RESULTS The aAPC propagated TIL at numbers equivalent to that found with PBMC feeders, whereas increasing the frequency of CD8 T-cell expansion with a comparable effector-memory phenotype. mRNA profiling revealed an upregulation of genes in the Wnt and stem-cell pathways with the aAPC. The aAPC platform did not skew clonal diversity, and CD8 T cells showed comparable antitumor function as those expanded with PBMC feeders. CONCLUSIONS TIL can be rapidly expanded with aAPC to clinical scale generating T cells with similar phenotypic and effector profiles as with PBMC feeders. These data support the clinical application of aAPC to manufacture TIL for the treatment of melanoma.
Collapse
|
45
|
Ott PA, Hodi FS, Buchbinder EI. Inhibition of Immune Checkpoints and Vascular Endothelial Growth Factor as Combination Therapy for Metastatic Melanoma: An Overview of Rationale, Preclinical Evidence, and Initial Clinical Data. Front Oncol 2015; 5:202. [PMID: 26442214 PMCID: PMC4585112 DOI: 10.3389/fonc.2015.00202] [Citation(s) in RCA: 188] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/31/2015] [Indexed: 01/29/2023] Open
Abstract
The role of angiogenesis as a mediator of immune regulation in the tumor microenvironment has recently come into focus. Furthermore, emerging evidence indicates that immunotherapy can lead to immune-mediated vasculopathy in the tumor, suggesting that the tumor vasculature may be an important interface between the tumor-directed immune response and the cancer itself. The advent of immune checkpoint inhibition as an effective immunotherapeutic strategy for many cancers has led to a better understanding of this interface. While the inhibition of angiogenesis through targeting of vascular endothelial growth factor (VEGF) has been used successfully for the treatment of cancer for many years, the mechanisms of its anti-tumor activity remain poorly understood. Initial studies of the complex relationship between angiogenesis, VEGF signaling and the immune system suggest that the combination of immune checkpoint blockade with angiogenesis inhibition has potential. While the majority of this work has been performed in metastatic melanoma, immunotherapy is rapidly showing promise in a broad range of malignancies and efforts to enhance immunotherapy will broadly impact the future of oncology. Here, we review the preclinical rationale and clinical investigations of combined angiogenesis inhibition and immunotherapy/immune checkpoint inhibition as a potentially promising combinatorial approach for cancer treatment.
Collapse
Affiliation(s)
- Patrick A Ott
- Department of Medical Oncology, Melanoma Disease Center, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Harvard Medical School , Boston, MA , USA ; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA
| | - F Stephen Hodi
- Department of Medical Oncology, Melanoma Disease Center, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Harvard Medical School , Boston, MA , USA ; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA
| | - Elizabeth I Buchbinder
- Department of Medical Oncology, Melanoma Disease Center, Center for Immuno-Oncology, Dana-Farber Cancer Institute, Harvard Medical School , Boston, MA , USA ; Department of Medicine, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
46
|
Tarhini AA, Zahoor H, Lin Y, Malhotra U, Sander C, Butterfield LH, Kirkwood JM. Baseline circulating IL-17 predicts toxicity while TGF-β1 and IL-10 are prognostic of relapse in ipilimumab neoadjuvant therapy of melanoma. J Immunother Cancer 2015; 3:39. [PMID: 26380086 PMCID: PMC4570556 DOI: 10.1186/s40425-015-0081-1] [Citation(s) in RCA: 296] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 07/10/2015] [Indexed: 01/31/2023] Open
Abstract
Background We evaluated candidate circulating serum cytokines, chemokines and growth factors in patients with locally/regionally advanced melanoma receiving neoadjuvant ipilimumab with toxicity and clinical outcome. Methods Patients were treated with ipilimumab (10 mg/kg IV every 3 weeks, 2 doses) before and after surgery. xMAP multiplex serum testing for 36 functionally selected cytokines and chemokines was performed at baseline and at six weeks (following ipilimumab). Based on our prior data, the association of IL-17 and immune related colitis was tested. Serum cytokines were divided into functional groups (Th1, Th2, Regulatory, Proinflammatory) and were assessed at baseline and week 6 using sparse-group Lasso modeling to assess the association of various cytokine groups with progression free survival (PFS). The linear combination of the cytokines/chemokines in this model was then used as a risk score and a Kaplan-Meier curve was generated to examine the association of the dichotomized score and PFS. Results Thirty-five patients were enrolled whose staging was: IIIB (3; N2b), IIIC (30; N2c, N3), IV (2). Median follow-up was 18 months. Among 33 evaluable patients, median PFS was 11 months (95 % CI = 6.2–19.2). IL-17 was found to correlate significantly with the incidence of grade 3 diarrhea/colitis when measured at baseline (p = 0.02) with a trend towards significance at 6 weeks (p = 0.06). In the modeling analysis, at baseline, the linear combination of 2 regulatory cytokines [TGF- β1 (ρ = 0.19) and IL-10 (ρ = -0.34)] was significantly associated with PFS (HR 2.66; p = 0.035). No significant correlations with clinical outcomes were found in examining the week 6 cytokines. Conclusions Baseline IL-17 level was significantly associated with the later development of severe diarrhea/colitis while the combination of baseline TGF- β1 and IL-10 levels were associated with therapeutic clinical outcome after neoadjuvant ipilimumab. These findings warrant further investigation and validation. Trial registration ClinicalTrials.gov Identifier NCT00972933.
Collapse
Affiliation(s)
- Ahmad A Tarhini
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - Haris Zahoor
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - Yan Lin
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - Usha Malhotra
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - Cindy Sander
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - Lisa H Butterfield
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| | - John M Kirkwood
- University of Pittsburgh Cancer Institute, UPMC Cancer Pavilion, 5150 Centre Avenue (555), Pittsburgh, PA 15232 USA
| |
Collapse
|
47
|
Schlößer HA, Theurich S, Shimabukuro-Vornhagen A, Holtick U, Stippel DL, von Bergwelt-Baildon M. Overcoming tumor-mediated immunosuppression. Immunotherapy 2015; 6:973-88. [PMID: 25341119 DOI: 10.2217/imt.14.58] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mechanisms of tumor-mediated immunosuppression have been described for several solid and hematological tumors. Tumors inhibit immune responses by attraction of immunosuppressive lymphocytic populations, secretion of immunosuppressive cytokines or expression of surface molecules, which inhibit immune responses by induction of anergy or apoptosis in tumor-infiltrating lymphocytes. This tumor-mediated immunosuppression represents a major obstacle to many immunotherapeutic or conventional therapeutic approaches. In this review we discuss how tumor-mediated immunosuppression interferes with different immunotherapeutic approaches and then give an overview of strategies to overcome it. Particular emphasis is placed on agents or approaches already transferred into clinical settings. Finally the success of immune checkpoint inhibitors targeting CTLA-4 or the PD-1 pathway highlights the enormous therapeutic potential of an effective overcoming of tumor-mediated immunosuppression.
Collapse
|
48
|
Raman R, Vaena D. Immunotherapy in Metastatic Renal Cell Carcinoma: A Comprehensive Review. BIOMED RESEARCH INTERNATIONAL 2015; 2015:367354. [PMID: 26161397 PMCID: PMC4486756 DOI: 10.1155/2015/367354] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 03/04/2015] [Indexed: 11/17/2022]
Abstract
Localized renal cell carcinoma (RCC) is often curable by surgery alone. However, metastatic RCC is generally incurable. In the 1990s, immunotherapy in the form of cytokines was the mainstay of treatment for metastatic RCC. However, responses were seen in only a minority of highly selected patients with substantial treatment-related toxicities. The advent of targeted agents such as vascular endothelial growth factor tyrosine kinase inhibitors VEGF-TKIs and mammalian target of rapamycin (mTOR) inhibitors led to a change in this paradigm due to improved response rates and progression-free survival, a better safety profile, and the convenience of oral administration. However, most patients ultimately progress with about 12% being alive at 5 years. In contrast, durable responses lasting 10 years or more are noted in a minority of those treated with cytokines. More recently, an improved overall survival with newer forms of immunotherapy in other malignancies (such as melanoma and prostate cancer) has led to a resurgence of interest in immune therapies in metastatic RCC. In this review we discuss the rationale for immunotherapy and recent developments in immunotherapeutic strategies for treating metastatic RCC.
Collapse
Affiliation(s)
- Rachna Raman
- Division of Hematology Oncology and Bone Marrow Transplantation, Department of Internal Medicine, University of Iowa Hospitals and Clinics, 200 Hawkins Drive C32GH, Iowa City, Iowa 52242, USA
| | - Daniel Vaena
- Division of Hematology Oncology and Bone Marrow Transplantation, Department of Internal Medicine, University of Iowa Hospitals and Clinics, 200 Hawkins Drive C32GH, Iowa City, Iowa 52242, USA
| |
Collapse
|
49
|
Yuan J, Zhou J, Dong Z, Tandon S, Kuk D, Panageas KS, Wong P, Wu X, Naidoo J, Page DB, Wolchok JD, Hodi FS. Pretreatment serum VEGF is associated with clinical response and overall survival in advanced melanoma patients treated with ipilimumab. Cancer Immunol Res 2015; 2:127-32. [PMID: 24778276 DOI: 10.1158/2326-6066.cir-13-0163] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Ipilimumab, an antibody that blocks CTL antigen 4 (CTLA-4), improves overall survival (OS) for patients with metastatic melanoma. Given its role in angiogenesis and immune evasion, serum VEGF levels were evaluated for association with clinical benefit in ipilimumab-treated patients. Sera were collected from 176 patients treated at 3 (n = 98) or 10 mg/kg (n = 68). The VEGF levels before treatment and at induction completion (week 12) were analyzed using the Meso Scale Discovery kit. The association of the levels of VEGF with clinical responses and OS were assessed using the Fisher exact and Kaplan-Meier log-rank tests. VEGF as a continuous variable was associated with OS (P = 0.002). Using 43 pg/mL as the cutoff pretreatment VEGF value defined by maximally selected log-rank statistics, pretreatment VEGF values correlated with clinical benefit at week 24 (P = 0.019; 159 patients evaluable). Pretreatment VEGF ≥ 43 pg/mL was associated with decreased OS (median OS 6.6 vs. 12.9 months, P = 0.006; 7.4 vs. 14.3 months, P = 0.037 for 3 mg/kg; and 6.2 vs. 10.9 months, P = 0.048 for 10 mg/kg). There was no correlation between VEGF changes and clinical outcome. Serum VEGF may be a predictive biomarker for ipilimumab treatment and is worthy of prospective investigation with various forms of immunologic checkpoint blockade.
Collapse
Affiliation(s)
- Jianda Yuan
- Authors' Affiliations Melanoma Disease Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Floros T, Tarhini AA. Anticancer Cytokines: Biology and Clinical Effects of Interferon-α2, Interleukin (IL)-2, IL-15, IL-21, and IL-12. Semin Oncol 2015; 42:539-48. [PMID: 26320059 DOI: 10.1053/j.seminoncol.2015.05.015] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Efforts over nearly four decades have focused on ways to use cytokines to manipulate the host immune response towards cancer cell recognition and eradication. Significant advances were achieved with interleukin-2 (IL-2) and interferon-α (IFN-α), primarily in the treatment of patients with melanoma and renal cell carcinoma. However, the utility of other cytokines showing promise in the preclinical setting has not been established largely because of toxicity, the complex functionality of each cytokine and the difficulty mimicking in preclinical models the human environment. Here, we review the basic biology and the clinical experiences with IFN-α, IL-2, IL-15, IL-21, and IL-12. We will also review ongoing clinical trials and discuss future directions including potential use of cytokines in combination with other effective immunotherapy approaches that have come of age in recent years.
Collapse
Affiliation(s)
- Theofanis Floros
- University of Pittsburgh Cancer Institute, Pittsburgh, PA; Athens Naval and Veterans Hospital, Pittsburgh, PA
| | - Ahmad A Tarhini
- University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Pittsburgh School of Medicine, Pittsburgh, PA.
| |
Collapse
|