scholarly journals Oncolytic viruses as a promising therapeutic strategy for hematological malignancies

2021 ◽  
Vol 139 ◽  
pp. 111573
Author(s):  
Chen Yang ◽  
Nanni Hua ◽  
Shufang Xie ◽  
Yi Wu ◽  
Lifeng Zhu ◽  
...  
2020 ◽  
Vol 28 (1) ◽  
pp. 159-183
Author(s):  
Vanessa Innao ◽  
Vincenzo Rizzo ◽  
Andrea Gaetano Allegra ◽  
Caterina Musolino ◽  
Alessandro Allegra

The use of viruses for tumour treatment has been imagined more than one hundred years ago, when it was reported that viral diseases were occasionally leading to a decrease in neoplastic lesions. Oncolytic viruses (OVs) seem to have a specific tropism for tumour cells. Previously, it was hypothesised that OVs’ antineoplastic actions were mainly due to their ability to contaminate, proliferate and destroy tumour cells and the immediate destructive effect on cells was believed to be the single mechanism of action of OVs’ action. Instead, it has been established that oncolytic viruses operate via a multiplicity of systems, including mutation of tumour milieu and a composite change of the activity of immune effectors. Oncolytic viruses redesign the tumour environment towards an antitumour milieu. The aim of our work is to evaluate the findings present in the literature about the use of OVs in the cure of haematological neoplastic pathologies such as multiple myeloma, acute and chronic myeloid leukaemia, and lymphoproliferative diseases. Further experimentations are essential to recognize the most efficient virus or treatment combinations for specific haematological diseases, and the combinations able to induce the strongest immune response.


Cancers ◽  
2020 ◽  
Vol 12 (5) ◽  
pp. 1295 ◽  
Author(s):  
Mizuho Sato-Dahlman ◽  
Christopher J. LaRocca ◽  
Chikako Yanagiba ◽  
Masato Yamamoto

Gene therapy with viral vectors has significantly advanced in the past few decades, with adenovirus being one of the most commonly employed vectors for cancer gene therapy. Adenovirus vectors can be divided into 2 groups: (1) replication-deficient viruses; and (2) replication-competent, oncolytic (OVs) viruses. Replication-deficient adenoviruses have been explored as vaccine carriers and gene therapy vectors. Oncolytic adenoviruses are designed to selectively target, replicate, and directly destroy cancer cells. Additionally, virus-mediated cell lysis releases tumor antigens and induces local inflammation (e.g., immunogenic cell death), which contributes significantly to the reversal of local immune suppression and development of antitumor immune responses (“cold” tumor into “hot” tumor). There is a growing body of evidence suggesting that the host immune response may provide a critical boost for the efficacy of oncolytic virotherapy. Additionally, genetic engineering of oncolytic viruses allows local expression of immune therapeutics, thereby reducing related toxicities. Therefore, the combination of oncolytic virus and immunotherapy is an attractive therapeutic strategy for cancer treatment. In this review, we focus on adenovirus-based vectors and discuss recent progress in combination therapy of adenoviruses with immunotherapy in preclinical and clinical studies.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. SCI-33-SCI-33
Author(s):  
Judy Lieberman

Abstract Abstract SCI-33 MicroRNAs regulate the response of the cell to environmental changes and developmental cues. microRNA expression is dysregulated in cancer – microRNA expression is generally reduced in cancer cells compared to normal tissue and individual microRNAs, termed oncomirs, that act as tumor suppressor genes or oncogenes are frequently aberrantly expressed in cancer and their expression can be linked to prognosis and response to therapy. Because microRNAs regulate cancer cell differentiation, proliferation, survival, and metastasis, manipulating microRNA function, either by mimicking or inhibiting miRNAs implicated in cancer, could provide a powerful therapeutic strategy to interfere with key pathways for cancer progression. This talk will explore some of the opportunities and obstacles to harnessing microRNA biology for cancer therapy. Disclosures: No relevant conflicts of interest to declare.


2018 ◽  
Vol 18 (2) ◽  
pp. 171-176 ◽  
Author(s):  
Min Liang

The oncolytic viruses now hold a promise of new therapeutic strategy for cancer. Its concept has inspired a wave of commercial research and development activities for the products of this category in China since 1998. The first commercialized oncolytic virus product in the world, Oncorine (H101), developed by Shanghai Sunway Biotech Co., Ltd since 1999, was approved by Chinese SFDA in November, 2005 for nasopharyngeal carcinoma in combination with chemotherapy after the phase III clinical trial, and finally acquired GMP certificate in August, 2006. This review introduces how Oncorine was successfully developed in China, and how the Chinese market responded after it was launched into the market in 2006.


2016 ◽  
Author(s):  
Carmelina Antonella Iannuzzi ◽  
Carmela Passaro ◽  
Silvia Boffo ◽  
Iris Maria Forte ◽  
Paola Indovina ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3941-3941 ◽  
Author(s):  
Nadine Hein ◽  
Sean O'Brien ◽  
Denis Drygin ◽  
Simon J. Harrison ◽  
Amit Khot ◽  
...  

Abstract Malignant transformation is commonly associated with dysregulated ribosome biogenesis and for more than 100 years, pathologists have utilized the increase in size and number of nucleoli, the site of ribosome biogenesis, as a marker for aggressive malignancies (Hein et al., Trends in Molecular Medicine, 2013). Recently we demonstrated that hyperactivated RNA Polymerase I (Pol I) transcription, a rate limiting step in ribosome biogenesis, can be specifically targeted by the small molecule inhibitor, CX-5461 (Drygin et al., Cancer Research, 2011; Bywater et al. Cancer Cell, 2012). When evaluated for its anti-proliferative activity against a panel of genetically diverse human cancer cell lines, those derived from p53 wild type hematological malignancies were the most sensitive (Drygin et al., Cancer Research, 2011). Evaluation of whether Pol I transcription inhibition could serve as therapeutic strategy in vivo revealed that CX-5461 administration significantly prolongs the overall survival of Em-Myc lymphoma bearing mice. This survival advantage was associated with rapid activation of the ribosomal protein (Rp)-MDM2-p53 nucleolar stress response (Deisenroth et al., Oncogene, 2010) and subsequent induction of tumour cell-specific p53-dependent apoptosis; importantly the wild type B-cell population was maintained and did not activate p53 pathways (Bywater et al. Cancer Cell, 2012). To explore the therapeutic potential of Pol I transcription inhibition via CX-5461 in hematological malignancies refractory to standard chemotherapy, we employed mouse models of highly aggressive MLL-driven Acute Myeloid Leukemia (AML) (MLL/AF9 + Nras and MLL/ENL + Nras) (Zuber et al., Genes Dev, 2009) and V*κ-Myc-driven Multiple Myeloma (MM) (Chesi et al. Cancer Cell, 2009). CX-5461 administration significantly increased the overall survival time of AML-bearing mice (MLL/ENL Nras median 17 days for vehicle vs 36 days for drug, P<0.0001 (Figure 1); MLL/AF9 Nras median 15 days for vehicle vs 23 days for drug, P 0.0009) without severe adverse effects. The extended survival of MLL-driven AML bearing mice was associated with acute activation of p53, induction of cell death and an aberrant G2/M cell cycle progression, which consequently delayed tumour progression as monitored by bioluminescence-imaging and white blood cell counts (WBC). In contrast to CX-5461, standard-therapy with a regime containing Cytarabine/Doxorubicin (5:3) failed to induce p53 in MLL/ENL Nras AML-bearing mice, resulting in a significantly lower survival advantage as compared to animals treated with CX-5461 therapy. Figure 1Figure 1. In ongoing experiments with MM-bearing mice, chronic CX-5461 administration robustly reduced the secretion of serum monoclonal Ig, as detected by serum protein electrophoresis (SPEP), and significantly prolonged the overall survival time (Figure 2) (V*κ-Myc median >160 days for drug vs, 103.5 days for vehicle, ongoing) demonstrating that CX-5461 exhibits potent anti-tumour activity in MM model. Analysis of the molecular mechanism responsible for the therapeutic benefit of CX-5461 in MM is ongoing but appears to involve both p53 mediated apoptosis and aberrant cell cycle progression. Figure 2Figure 2. These data demonstrate that hyperactivated Pol I transcription can be successfully targeted through small molecule inhibitors to treat models of human AML and MM that are refractory to standard therapy. Based on these and previously published results (Bywater et al., Cancer Cell, 2012), we have recently initiated a first-in-human, phase I, dose escalation clinical trial of this first-in-class drug, CX-5461 in patients with advanced hematological malignancies. The toxicity, response, pharmacokinetic and co-relative data from this trial will offer valuable insights as to whether inhibition of ribosome synthesis might offer a new non-genotoxic therapeutic approach in the fight against cancers, which are highly refractory to standard therapies. Disclosures: O'Brien: Senhwa Biosciences, Inc: Employment.


2012 ◽  
Vol 16 (7) ◽  
pp. 729-742 ◽  
Author(s):  
Alberto M Martelli ◽  
Francesca Chiarini ◽  
Camilla Evangelisti ◽  
Andrea Ognibene ◽  
Daniela Bressanin ◽  
...  

2020 ◽  
Vol 19 (1) ◽  
Author(s):  
Bin Zhang ◽  
Ping Cheng

Abstract As a promising therapeutic strategy, oncolytic virotherapy has shown potent anticancer efficacy in numerous pre-clinical and clinical trials. Oncolytic viruses have the capacity for conditional-replication within carcinoma cells leading to cell death via multiple mechanisms, including direct lysis of neoplasms, induction of immunogenic cell death, and elicitation of innate and adaptive immunity. In addition, these viruses can be engineered to express cytokines or chemokines to alter tumor microenvironments. Combination of oncolytic virotherapy with other antitumor therapeutic modalities, such as chemotherapy and radiation therapy as well as cancer immunotherapy can be used to target a wider range of tumors and promote therapeutic efficacy. In this review, we outline the basic biological characteristics of oncolytic viruses and the underlying mechanisms that support their use as promising antitumor drugs. We also describe the enhanced efficacy attributed to virotherapy combined with other drugs for the treatment of cancer.


Blood ◽  
2017 ◽  
Vol 129 (1) ◽  
pp. 88-99 ◽  
Author(s):  
Changchun Deng ◽  
Mark R. Lipstein ◽  
Luigi Scotto ◽  
Xavier O. Jirau Serrano ◽  
Michael A. Mangone ◽  
...  

Key Points A novel PI3Kδ inhibitor TGR-1202 synergizes with proteasome inhibitor carfilzomib by silencing c-Myc in preclinical models of lymphoma. The unique activity of TGR-1202 as a single agent and in combination with carfilzomib is driven by an unexpected activity targeting CK1ε.


Sign in / Sign up

Export Citation Format

Share Document