1
|
Sheth RA, Wehrenberg-Klee E, Patel SP, Brock KK, Fotiadis N, de Baère T. Intratumoral Injection of Immunotherapeutics: State of the Art and Future Directions. Radiology 2024; 312:e232654. [PMID: 39078294 PMCID: PMC11294769 DOI: 10.1148/radiol.232654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 02/17/2024] [Accepted: 02/28/2024] [Indexed: 07/31/2024]
Abstract
Systemic immunotherapies have led to tremendous progress across the cancer landscape. However, several challenges exist, potentially limiting their efficacy in the treatment of solid tumors. Direct intratumoral injection can increase the therapeutic index of immunotherapies while overcoming many of the barriers associated with systemic administration, including limited bioavailability to tumors and potential systemic safety concerns. However, challenges remain, including the lack of standardized approaches for administration, issues relating to effective drug delivery, logistical hurdles, and safety concerns specific to this mode of administration. This article reviews the biologic rationale for the localized injection of immunotherapeutic agents into tumors. It also addresses the existing limitations and practical considerations for safe and effective implementation and provide recommendations for optimizing logistics and treatment workflows. It also highlights the critical role that radiologists, interventional radiologists, and medical physicists play in intratumoral immunotherapy with respect to target selection, image-guided administration, and response assessment.
Collapse
Affiliation(s)
- Rahul A. Sheth
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Eric Wehrenberg-Klee
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Sapna P. Patel
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Kristy K. Brock
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Nicos Fotiadis
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| | - Thierry de Baère
- From the Departments of Interventional Radiology (R.A.S.), Melanoma
Medical Oncology (S.P.P.), and Imaging Physics (K.K.B.), University of Texas MD
Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; Department of
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K.); Department of
Radiology, Royal Marsden Hospital, London, England (N.F.); and Department of
Interventional Radiology, Institut de Cancérologie Gustave Roussy,
Villejuif, France (T.d.B.)
| |
Collapse
|
2
|
Noval Rivas M, Rosser CJ, Arditi M. Rationale for Randomized Clinical Trials Investigating the Potential of BCG Vaccination in Preventing COVID-19 Infection. Bladder Cancer 2021; 7:121-131. [PMID: 38994533 PMCID: PMC11181712 DOI: 10.3233/blc-201529] [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: 12/11/2020] [Accepted: 02/04/2021] [Indexed: 11/15/2022]
Abstract
Despite the implementation of mitigation measures, Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is still spreading worldwide, and has caused more than 1 million deaths so far. Although recent reports indicate that three vaccine candidates are effective against SARS-CoV-2, more time is needed to generate enough doses for the general population. Meanwhile, frontline healthcare workers are at high risk of SARS-CoV-2 exposure. To avoid collapse of the medical care system, there is a need to develop novel approaches to limit SARS-CoV-2 spread. Through a process called trained immunity, the Bacillus Calmette-Guerin (BCG) vaccine boosts the action of innate immune cells, resulting in a nonspecific reduction in the incidence of viral infections. Due to this immunomodulatory action, the BCG vaccine is currently used as a therapeutic in bladder cancer. Data collected from epidemiological and observational studies indicate that BCG vaccination might provide protection against COVID-19. While these observations do not provide evidence of causality and are limited by cofounding and intrinsic biases, it is crucial to explore the hypothesis that BCG vaccination may provide a nonspecific innate immune boost and therefore protect against COVID-19 in randomized controlled clinical trials, particularly for people at higher risk of developing COVID-19, such as frontline healthcare workers.
Collapse
Affiliation(s)
- Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Charles J. Rosser
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Moshe Arditi
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Infectious and Immunologic Diseases Research Center (IIDRC) and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
3
|
Aspatwar A, Gong W, Wang S, Wu X, Parkkila S. Tuberculosis vaccine BCG: the magical effect of the old vaccine in the fight against the COVID-19 pandemic. Int Rev Immunol 2021; 41:283-296. [PMID: 33960271 PMCID: PMC8108189 DOI: 10.1080/08830185.2021.1922685] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Bacillus Calmette-Guérin (BCG) is a live attenuated M. bovis vaccine that was developed about 100 years ago by Albert Calmette and Camille Guérin. Many countries have been using the vaccine for decades against tuberculosis (TB). The World Health Organization (WHO) recommends a single dose of BCG for infants in TB endemic as well as leprosy high risk countries, and globally almost 130 million infants are vaccinated yearly. The role of BCG is well known in reducing neonatal and childhood death rates. Epidemiological and retrospective cross-sectional studies demonstrated that the BCG vaccination protects the children against respiratory tract infections and lowers the risk of malaria in children. In addition, BCG enhances IFN-γ and IL-10 levels, thus providing immunity against respiratory tract infection even in elderly people. The BCG is also known to provide nonspecific innate immunity against viruses and parasites, through an innate immune mechanism termed ‘trained immunity’ and is defined as the immunological recall of the innate immune system by epigenetic reprogramming. Based on these studies it is suggested that the BCG has the potential to act as a protective agent against COVID-19. Further proven safety records of BCG in humans, its adjuvant activity and low-cost manufacturing make it an attractive option to stop the pandemic and reduce the COVID-19 related mortality. In this review we discuss the heterologous effects of BCG, induction of trained immunity and its implication in development of a potential vaccine against COVID-19 pandemic.
Collapse
Affiliation(s)
- Ashok Aspatwar
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Wenping Gong
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, the 8th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shuyong Wang
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, the 8th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xueqiong Wu
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, the 8th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Seppo Parkkila
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Fimlab Laboratories Ltd, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
4
|
Klarquist JS, Janssen EM. Melanoma-infiltrating dendritic cells: Limitations and opportunities of mouse models. Oncoimmunology 2021; 1:1584-1593. [PMID: 23264904 PMCID: PMC3525613 DOI: 10.4161/onci.22660] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The infiltration of melanoma lesions by dendritic cells (DCs) has been suggested to play a tumorigenic role due to the capacity of DCs to induce tumor tolerance and promote angiogenesis as well as metastasis. However, it has also been shown that tumor-infiltrating DCs (TIDCs) induce antitumor responses and hence may be targeted in cost-effective therapeutic approaches to obtain patient-specific DCs that present relevant tumor antigens, without the need for ex vivo DC expansion or tumor antigen identification. Unfortunately, little is known about the composition, nature and function of TIDCs found in human melanoma. The development of mouse melanoma models has greatly contributed to the molecular understanding of melanoma immunology in mice, but many questions on TIDCs remain unanswered. Here, we discuss current knowledge about melanoma TIDCs in various mouse models with regard to their translational potential and clinical relevance.
Collapse
Affiliation(s)
- Jared S Klarquist
- Division of Cellular and Molecular Immunology; Cincinnati Children's Hospital Research Foundation; University of Cincinnati College of Medicine; Cincinnati, OH USA
| | | |
Collapse
|
5
|
Kremenovic M, Schenk M, Lee DJ. Clinical and molecular insights into BCG immunotherapy for melanoma. J Intern Med 2020; 288:625-640. [PMID: 32128919 DOI: 10.1111/joim.13037] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/23/2019] [Accepted: 01/24/2020] [Indexed: 01/25/2023]
Abstract
The incidence of cutaneous melanoma and the mortality rate of advanced melanoma patients continue to rise globally. Despite the recent success of immunotherapy including ipilimumab and pembrolizumab checkpoint inhibitors, a large proportion of patients are refractory to such treatment modalities. The application of mycobacteria such as Bacillus Calmette-Guérin (BCG) in the treatment of various malignancies, including cutaneous melanoma, has been clearly demonstrated after almost a century of observations and experimentation. Intralesional BCG (IL-BCG) immunotherapy is a highly efficient and cost-effective treatment option for inoperable stage III in-transit melanoma, as recommended in the National Comprehensive Cancer Network Guidelines. IL-BCG has shown great efficacy in the regression of directly injected metastatic melanoma lesions, as well as distal noninjected nodules in immunocompetent patients. Clinical and preclinical studies have shown that BCG serves as a strong immune modulator, inducing the recruitment of various immune cells that contribute to antitumour immunity. However, the specific mechanism of BCG-mediated tumour immunity remains poorly understood. Comparative genome analyses have revealed that different BCG strains exhibit distinct immunological activity and virulence, which might impact the therapeutic response and clinical outcome of patients. In this review, we discuss the immunostimulatory potential of different BCG substrains and highlight clinical studies utilizing BCG immunotherapy for the treatment of cutaneous melanoma. Furthermore, the review focuses on the cellular and molecular mechanisms of the BCG-induced immune responses of both the innate and adaptive arms of the immune system. Furthermore, the review discussed the administration of BCG as a monotherapy or in combination with other immunotherapeutic or chemotherapeutic agents.
Collapse
Affiliation(s)
- M Kremenovic
- From the, Institute of Pathology, Experimental Pathology, Universitat Bern, Bern, Switzerland
| | - M Schenk
- From the, Institute of Pathology, Experimental Pathology, Universitat Bern, Bern, Switzerland
| | - D J Lee
- Division of Dermatology, Department of Medicine, The Lundquist Institute, Los Angeles, CA, USA
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
6
|
Squarzanti DF, Zavattaro E, Pizzimenti S, Amoruso A, Savoia P, Azzimonti B. Non-Melanoma Skin Cancer: news from microbiota research. Crit Rev Microbiol 2020; 46:433-449. [PMID: 32692305 DOI: 10.1080/1040841x.2020.1794792] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Recently, research has been deeply focusing on the role of the microbiota in numerous diseases, either affecting the skin or other organs. What it is well established is that its dysregulation promotes several cutaneous disorders (i.e. psoriasis and atopic dermatitis). To date, little is known about its composition, mediators and role in the genesis, progression and response to therapy of Non-Melanoma Skin Cancer (NMSC). Starting from a bibliographic study, we classified the selected articles into four sections: i) normal skin microbiota; ii) in vitro study models; iii) microbiota and NMSC and iv) probiotics, antibiotics and NMSC. What has emerged is how skin microflora changes, mainly represented by increases of Staphylococcus aureus, Streptococcus pyogenes and Pseudomonas aeruginosa strains, modifications in the mutual quantity of β-Human papillomavirus genotypes, of Epstein Barr Virus and Malassezia or candidiasis, may contribute to the induction of a state of chronic self-maintaining inflammation, leading to cancer. In this context, the role of S. aureus and that of specific antimicrobial peptides look to be prominent. Moreover, although antibiotics may contribute to carcinogenesis, due to their ability to influence the microbiota balance, specific probiotics, such as Lacticaseibacillus rhamnosus GG, Lactobacillus johnsonii NCC 533 and Bifidobacteria spp., may be protective.
Collapse
Affiliation(s)
- Diletta Francesca Squarzanti
- Department of Health Sciences (DiSS), University of Piemonte Orientale (UPO), Novara, Italy.,Center for Translational Research on Autoimmune and Allergic Diseases (CAAD), DiSS, UPO, Novara, Italy
| | - Elisa Zavattaro
- Department of Translational Medicine (DiMeT), UPO, Novara, Italy
| | - Stefania Pizzimenti
- Department of Clinical and Biological Sciences (DSCB), University of Turin, Turin, Italy
| | | | - Paola Savoia
- Department of Health Sciences (DiSS), University of Piemonte Orientale (UPO), Novara, Italy
| | - Barbara Azzimonti
- Department of Health Sciences (DiSS), University of Piemonte Orientale (UPO), Novara, Italy.,Center for Translational Research on Autoimmune and Allergic Diseases (CAAD), DiSS, UPO, Novara, Italy
| |
Collapse
|
7
|
Vidovic D, Giacomantonio C. Insights into the Molecular Mechanisms Behind Intralesional Immunotherapies for Advanced Melanoma. Cancers (Basel) 2020; 12:cancers12051321. [PMID: 32455916 PMCID: PMC7281646 DOI: 10.3390/cancers12051321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/17/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
The incidence of cutaneous melanoma, a highly malignant skin cancer, is increasing yearly. While surgical removal of the tumor is the mainstay of treatment for patients with locally confined disease, those with metastases face uncertainty when it comes to their treatment. As melanoma is a relatively immunogenic cancer, current guidelines suggest using immunotherapies that can rewire the host immune response to target melanoma tumor cells. Intralesional therapy, where immunomodulatory agents are injected directly into the tumor, are an emerging aspect of treatment for in-transit melanoma because of their ability to mitigate severe off-target immune-related adverse events. However, their immunomodulatory mechanisms are poorly understood. In this review, we will summarize and discuss the different intralesional therapies for metastatic melanoma with respect to their clinical outcomes and immune molecular mechanisms.
Collapse
|
8
|
Benitez MLR, Bender CB, Oliveira TL, Schachtschneider KM, Collares T, Seixas FK. Mycobacterium bovis BCG in metastatic melanoma therapy. Appl Microbiol Biotechnol 2019; 103:7903-7916. [PMID: 31402426 DOI: 10.1007/s00253-019-10057-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/22/2019] [Accepted: 07/26/2019] [Indexed: 12/13/2022]
Abstract
Melanoma is the most aggressive form of skin cancer, with a high mortality rate and with 96,480 new cases expected in 2019 in the USS. BRAFV600E, the most common driver mutation, is found in around 50% of melanomas, contributing to tumor growth, angiogenesis, and metastatic progression. Dacarbazine (DTIC), an alkylate agent, was the first chemotherapeutic agent approved by the US Food and Drug Administration (FDA) used as a standard treatment. Since then, immunotherapies have been approved for metastatic melanoma (MM) including ipilimumab and pembrolizumab checkpoint inhibitors that help decrease the risk of progression. Moreover, Mycobacterium bovis Bacillus Calmette-Guerin (BCG) serves as an adjuvant therapy that induces the recruitment of natural killer NK, CD4+, and CD8+ T cells and contributes to antitumor immunity. BCG can be administered in combination with chemotherapeutic and immunotherapeutic agents and can be genetically manipulated to produce recombinant BCG (rBCG) strains that express heterologous proteins or overexpress immunogenic proteins, increasing the immune response and improving patient survival. In this review, we highlight several studies utilizing rBCG immunotherapy for MM in combination with other therapeutic agents.
Collapse
Affiliation(s)
- Martha Lucia Ruiz Benitez
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Camila Bonnemann Bender
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Thaís Larré Oliveira
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Kyle M Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, USA.,Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA.,National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Tiago Collares
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil
| | - Fabiana Kömmling Seixas
- Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Rio Grande do Sul, Brazil.
| |
Collapse
|
9
|
Couto GK, Segatto NV, Oliveira TL, Seixas FK, Schachtschneider KM, Collares T. The Melding of Drug Screening Platforms for Melanoma. Front Oncol 2019; 9:512. [PMID: 31293965 PMCID: PMC6601395 DOI: 10.3389/fonc.2019.00512] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/28/2019] [Indexed: 12/30/2022] Open
Abstract
The global incidence of cancer is rising rapidly and continues to be one of the leading causes of death in the world. Melanoma deserves special attention since it represents one of the fastest growing types of cancer, with advanced metastatic forms presenting high mortality rates due to the development of drug resistance. The aim of this review is to evaluate how the screening of drugs and compounds for melanoma has been performed over the last seven decades. Thus, we performed literature searches to identify melanoma drug screening methods commonly used by research groups during this timeframe. In vitro and in vivo tests are essential for the development of new drugs; however, incorporation of in silico analyses increases the possibility of finding more suitable candidates for subsequent tests. In silico techniques, such as molecular docking, represent an important and necessary first step in the screening process. However, these techniques have not been widely used by research groups to date. Our research has shown that the vast majority of research groups still perform in vitro and in vivo tests, with emphasis on the use of in vitro enzymatic tests on melanoma cell lines such as SKMEL and in vivo tests using the B16 mouse model. We believe that the union of these three approaches (in silico, in vitro, and in vivo) is essential for improving the discovery and development of new molecules with potential antimelanoma action. This workflow would provide greater confidence and safety for preclinical trials, which will translate to more successful clinical trials and improve the translatability of new melanoma treatments into clinical practice while minimizing the unnecessary use of laboratory animals under the principles of the 3R's.
Collapse
Affiliation(s)
- Gabriela Klein Couto
- Research Group in Molecular and Cellular Oncology, Postgraduate Program in Biochemistry and Bioprospecting, Cancer Biotechnology Laboratory, Center for Technological Development, Federal University of Pelotas, Pelotas, Brazil
| | - Natália Vieira Segatto
- Biotechnology Graduate Program, Molecular and Cellular Oncology Research Group, Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Brazil
| | - Thaís Larré Oliveira
- Biotechnology Graduate Program, Molecular and Cellular Oncology Research Group, Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Brazil
| | - Fabiana Kömmling Seixas
- Biotechnology Graduate Program, Molecular and Cellular Oncology Research Group, Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Brazil
| | - Kyle M Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, United States.,Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL, United States.,National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Tiago Collares
- Biotechnology Graduate Program, Molecular and Cellular Oncology Research Group, Laboratory of Cancer Biotechnology, Technology Development Center, Federal University of Pelotas, Pelotas, Brazil
| |
Collapse
|
10
|
Molecular background of skin melanoma development and progression: therapeutic implications. Postepy Dermatol Alergol 2019; 36:129-138. [PMID: 31320844 PMCID: PMC6627250 DOI: 10.5114/ada.2019.84590] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 02/18/2018] [Indexed: 12/19/2022] Open
Abstract
Melanoma is the most aggressive skin cancer with an increasing number of cases worldwide and curable mostly in its early stage. The improvement in patients' survival in advanced melanoma has been achieved only recently, due to development of new biological drugs for targeted therapies and immunotherapy. Further progress in the treatment of melanoma is clearly dependent on the better understanding of its complex biology. This review describes the most important molecular mechanisms and genetic events underlying skin melanoma development and progression, depicts the way of action of newly developed drugs and indicates new potential therapeutic targets.
Collapse
|
11
|
Addressing the Adult Soft Tissue Sarcoma Microenvironment with Intratumoral Immunotherapy. Sarcoma 2018; 2018:9305294. [PMID: 30158830 PMCID: PMC6109466 DOI: 10.1155/2018/9305294] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/16/2018] [Indexed: 02/07/2023] Open
Abstract
Sarcoma is comprised of a heterogeneous group of tumors originating from the mesenchyme. Sarcoma is also the first tumor that responded to immunotherapeutic agents often termed as “Coley's toxins.” However, immunotherapy is yet to establish its presence in sarcomas. Complex interactions between tumor and immune cells in the tumor microenvironment play a crucial role in response to immunotherapy. There is a dynamic equilibrium created by the immune cells infiltrating the tumor, and this forms the basis of tumor evasion. Manipulating the intratumoral microenvironment will help overcome tumor evasion.
Collapse
|
12
|
Lardone RD, Chan AA, Lee AF, Foshag LJ, Faries MB, Sieling PA, Lee DJ. Mycobacterium bovis Bacillus Calmette-Guérin Alters Melanoma Microenvironment Favoring Antitumor T Cell Responses and Improving M2 Macrophage Function. Front Immunol 2017; 8:965. [PMID: 28848560 PMCID: PMC5554507 DOI: 10.3389/fimmu.2017.00965] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/28/2017] [Indexed: 01/07/2023] Open
Abstract
Intralesional Mycobacterium bovis bacillus Calmette-Guérin (BCG) has long been a relatively inexpensive therapy for inoperable cutaneous metastatic melanoma (CMM), although intralesional BCG skin mechanisms remain understudied. We analyzed intralesional BCG-treated CMM lesions combined with in vitro studies to further investigate BCG-altered pathways. Since macrophages play a pivotal role against both cancer and mycobacterial infections, we hypothesized BCG regulates macrophages to promote antitumor immunity. Tumor-associated macrophages (M2) infiltrate melanomas and impair antitumor immunity. BCG-treated, in vitro-polarized M2 (M2-BCG) showed transcriptional changes involving inflammation, immune cell recruitment, cross talk, and activation pathways. Mechanistic network analysis indicated M2-BCG potential to improve interferon gamma (IFN-γ) responses. Accordingly, frequency of IFN-γ-producing CD4+ T cells responding to M2-BCG vs. mock-treated M2 increased (p < 0.05). Moreover, conditioned media from M2-BCG vs. M2 elevated the frequency of granzyme B-producing CD8+ tumor-infiltrating lymphocytes (TILs) facing autologous melanoma cell lines (p < 0.01). Furthermore, transcriptome analysis of intralesional BCG-injected CMM relative to uninjected lesions showed immune function prevalence, with the most enriched pathways representing T cell activation mechanisms. In vitro-infected MM-derived cell lines stimulated higher frequency of IFN-γ-producing TIL from the same melanoma (p < 0.05). Our data suggest BCG favors antitumor responses in CMM through direct/indirect effects on tumor microenvironment cell types including macrophages, T cells, and tumor itself.
Collapse
Affiliation(s)
- Ricardo D Lardone
- Dirks/Dougherty Laboratory for Cancer Research, Department of Translational Immunology, John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Alfred A Chan
- Dirks/Dougherty Laboratory for Cancer Research, Department of Translational Immunology, John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States.,Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Agnes F Lee
- Dirks/Dougherty Laboratory for Cancer Research, Department of Translational Immunology, John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Leland J Foshag
- Division of Surgical Oncology, John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Mark B Faries
- Melanoma Research Program, John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Peter A Sieling
- Dirks/Dougherty Laboratory for Cancer Research, Department of Translational Immunology, John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Delphine J Lee
- Dirks/Dougherty Laboratory for Cancer Research, Department of Translational Immunology, John Wayne Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, United States.,Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States
| |
Collapse
|
13
|
Leventhal JS, Odell ID, Imaeda S, Maverakis E, King BA. Treatment of melanoma in-transit metastases with combination intralesional interleukin-2, topical imiquimod, and tretinoin 0.1% cream. JAAD Case Rep 2016; 2:114-6. [PMID: 27051847 PMCID: PMC4810288 DOI: 10.1016/j.jdcr.2016.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Jonathan S. Leventhal
- Yale University School of Medicine, Department of Dermatology, New Haven, Connecticut
| | - Ian D. Odell
- Yale University School of Medicine, Department of Dermatology, New Haven, Connecticut
| | - Suguru Imaeda
- Yale University School of Medicine, Department of Dermatology, New Haven, Connecticut
- Department of Veterans Affairs, West Haven, Connecticut
| | | | - Brett A. King
- Yale University School of Medicine, Department of Dermatology, New Haven, Connecticut
- Department of Veterans Affairs, West Haven, Connecticut
- Correspondence to: Brett A. King, MD, PhD, Yale University School of Medicine, Department of Dermatology, 15 York Street, LMP 5040, New Haven, CT 06510.Yale University School of MedicineDepartment of Dermatology15 York StreetLMP 5040New HavenCT06510
| |
Collapse
|
14
|
Electrochemotherapy for the management of melanoma skin metastasis: a review of the literature and possible combinations with immunotherapy. Arch Dermatol Res 2014; 306:521-6. [DOI: 10.1007/s00403-014-1462-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 01/09/2014] [Accepted: 03/03/2014] [Indexed: 02/03/2023]
|
15
|
Abstract
Surgical excision is the treatment of choice for primary melanomas and radiation therapy is the accepted alternative for the subset of lesions not amenable to surgery. With the recent rise in melanoma incidence, especially in the elderly, there are a growing number of cases that are neither amenable to surgery nor radiation therapy. In this article, we review pharmacotherapeutic approaches to microinvasive melanoma (invasive radial growth phase melanoma) that might be considered in such circumstances. There are no approved drugs for the treatment of primary melanoma and randomized controlled trials with 5 or more years of follow-up have not been performed. The limited studies and numerous case series in the literature on pharmacologic treatment of primary melanoma have focused on topical therapies. Accordingly, we provide a review of the potential pharmacotherapeutic agents in the treatment of microinvasive melanoma by extrapolating from the available limited literature on the use of fluorouracil, azelaic acid, retinoic acid derivatives, interferon (IFN)-α, imiquimod, and other agents for melanoma in situ, invasive melanoma, and epidermotropic melanoma metastases. Our review indicates that topical fluorouracil and tretinoin are not effective as single agents. The efficacy of azelaic acid, tazarotene, cidofovir, and intralesional IFN-α, interleukin-2, and IFN-β is undefined. Imiquimod is the most studied and promising agent; however, optimal dosage, therapeutic regimen, and survival rates are unknown. In the face of a growing demand for non-surgical treatments, formal clinical trials are needed to ascertain the role of pharmacotherapeutic agents in the treatment of microinvasive melanoma.
Collapse
Affiliation(s)
- Elizabeth A Quigley
- Dermatology Service, Memorial Sloan Kettering Cancer Center, 136 Mountain View Blvd, Basking Ridge, NJ, USA.
| | | |
Collapse
|
16
|
Albertini MR, Hank JA, Gadbaw B, Kostlevy J, Haldeman J, Schalch H, Gan J, Kim K, Eickhoff J, Gillies SD, Sondel PM. Phase II trial of hu14.18-IL2 for patients with metastatic melanoma. Cancer Immunol Immunother 2012; 61:2261-71. [PMID: 22678096 DOI: 10.1007/s00262-012-1286-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 05/17/2012] [Indexed: 01/12/2023]
Abstract
Phase I testing of the hu14.18-IL2 immunocytokine in melanoma patients showed immune activation, reversible toxicities, and a maximal tolerated dose of 7.5 mg/m(2)/day. In this phase II study, 14 patients with measurable metastatic melanoma were scheduled to receive hu14.18-IL2 at 6 mg/m(2)/day as 4-h intravenous infusions on Days 1, 2, and 3 of each 28 day cycle. Patients with stable disease (SD) or regression following cycle 2 could receive two additional treatment cycles. The primary objective was to evaluate antitumor activity and response duration. Secondary objectives evaluated adverse events and immunologic activation. All patients received two cycles of treatment. One patient had a partial response (PR) [1 PR of 14 patients = response rate of 7.1 %; confidence interval, 0.2-33.9 %], and 4 patients had SD and received cycles 3 and 4. The PR and SD responses lasted 3-4 months. All toxicities were reversible and those resulting in dose reduction included grade 3 hypotension (2 patients) and grade 2 renal insufficiency with oliguria (1 patient). Patients had a peripheral blood lymphocytosis on Day 8 and increased C-reactive protein. While one PR in 14 patients met protocol criteria to proceed to stage 2 and enter 16 additional patients, we suspended stage 2 due to limited availability of hu14.18-IL2 at that time and the brief duration of PR and SD. We conclude that subsequent testing of hu14.18-IL2 should involve melanoma patients with minimal residual disease based on compelling preclinical data and the confirmed immune activation with some antitumor activity in this study.
Collapse
Affiliation(s)
- Mark R Albertini
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|