kabilan A, Lakshmi L, Priyadarshoni P. S. Potential of Genetically- Modified Measles Virus as a Treatment Modality for Carcinoma – A Review. Biomed Pharmacol J 2018;11(2).
Manuscript received on :26 April 2018
Manuscript accepted on :01 June 2018
Published online on: 27-06-2018
Plagiarism Check: Yes
How to Cite    |   Publication History
Views Views: (Visited 692 times, 1 visits today)   Downloads PDF Downloads: 453

Anirudha kabilan1, Lakshmiand Pavithra Priyadarshoni S1

1Saveetha Institute of medical and technical sciences. Chennai, India.

2Department of pathology Saveetha  institute of medical and technical sciences. Chennai, India.

Corresponding Author E-mail: drpavithra88@gmail.com

DOI : https://dx.doi.org/10.13005/bpj/1448

Abstract

Malignancy is a disease in which cell division is uncontrolled and prognosis is often poor. Despite recent advances in the felid of medicine the life expectancy after the diagnosis of advanced stages of cancers  has high mortality rates  . The traditional methods of treatment have low curative effects and high risk of side effects. Further the possibility of re-occurrence is not completely eliminated by any of the conventional methods of treatment. Thus, a technique that affects only the tumour cells without leaving behind any cancer initiator cells must be deviced.  Recently genetically modified variants of measles virus were used to cure multiple myeloma .The idea to use of measles virus dates back to 1950’s.Constant research has lead  the advent  of a branch known as oncolytic virotheraphy . Precise targeting of cancer cells is one of the dominant advantages of cancer therapy through virus and it can be achieved in multiple manners. A few viruses such as   exclusively  replicating mumps virus, moloney leukemia virus, parvoviruses, reovirus, newcastle disease virus  have a natural preference for malignant  cells, whereas vesicular stomatitis adenovirus, virus, measles, vaccinia and herpes simplex virus can be adapted or engineered to make them cancer-specific.

Keywords

Genetically Modified; Measles; Oncolytic; Virus; Virotherapy

Download this article as: 
Copy the following to cite this article:

kabilan A, Lakshmi L, Priyadarshoni P. S. Potential of Genetically- Modified Measles Virus as a Treatment Modality for Carcinoma – A Review. Biomed Pharmacol J 2018;11(2).

Copy the following to cite this URL:

kabilan A, Lakshmi L, Priyadarshoni P. S. Potential of Genetically- Modified Measles Virus as a Treatment Modality for Carcinoma – A Review. Biomed Pharmacol J 2018;11(2). Available from: http://biomedpharmajournal.org/?p=21003

Introduction

For more than a century, viruses have been considered as potent experimental agents to eliminate or regress neoplastic growths.1 A clear perspective about viruses increased in the 1950s and 1960s, immensely due to the development of cell and tissue culture systems which allowed vivo virus breeding.2,3 An early approach for the cure of cancer was through a toxin commonly known as the Colley’s toxin. The toxin contained killed bacteria and proteins. Though Colley’s toxin was not proven to be beneficial.4 Later scientist tried to use infectious agents for the cure of cancer. In 1950’s it was noticed that West Nile virus had tumour shrinking properties. West Nile virus had the risk of causing or developing a disease which is known as West Nile encephalitis. Therefore clinical trials had to come to an end. The history of oncolytic virotheraphy dates back to the 12th century that documented spontaneous regression of haematological cancers after wild measles infection. Over  the past fifty years, viruses have been investigated wiintensity and their  biology is now appreciated more comprehensively than that of any other organism in nature. These efforts have led to better understanding of genomes and proteins, their physical structures, their replication cycles and pathogenetic strategies futher the ability to regulate  their genomes have been deviced.5 After constant research, oncolytic viruses were engineered. Various types of viruses like herpes virus, influenza virus, pox virus are being tested for their oncolytic properties.6  The oldest vaccine used for the eradication of small pox is being researched for its oncolytic properties.7 The modernised rein of oncolytic virotherapy, in which virus genomes are tailored  to enhance their anti-tumor specificity, can be traced to a 1991 publication in which a thymidine kinase (TK)-negative herpes simplex virus (HSV) with attenuated neurovirulence was shown to be active in a murine.Presently the most cumbersome task is to find out the right kind of virus for destruction of particular type of tumour cells. Recently the cure of multiple myeloma was brought about by injecting genetically modified variants of measles virus. This progress brought the field of oncolytic virotheraphy into lime light.

Oncolytic Virotheraphy

Viruses can specifically infect and lyse the tumour cells.8 The basis for oncolysis rests on the below factors.

Wild strains that affect the cancer cells.

Attenuated mutants of human virus strains.

Viruses attenuated by culturing techniques.

The viral genes perform as tumour toxic agents and the capsids acts as vehicles.Oncolytic virus acquire  their distinctive feature either by exploiting the cell surface receptors or intracellular gene aberration which are over expressed in cancer cells.8 One of the greatest advantages of oncolytic virotheraphy is the ability to engineer the virus according to the outcomes of clinical trials. Cancer cells show altered cell physiology like insensitivity to inhibitory growth signals, extensive replicative potential, tissue invasion and metastatis and sustained angiogenisis. These alterations in cell physiology make selective replication of the virus possible.9 Cancer targeting techniques of virus can be achieved by two approaches either by deleting the viral genes required for virus replication in normal cells or by using tumour specific promoters for viral genes.10 Experimentts performed with other oncolytic virus like reovirus and herpes virus exhibit  that cyclophosphamide decrease the innate immune responses,  extend viral gene expression and proliferation, and improve oncolytic effect. Alternate mechanisms to target cancer cells is to distinctively erase off  the  undesirable tropism. This is achieved by specifically constructing the virus for various specified target organs in their genomes to facilitate the selective blocking of the virus’s  life cycle  in the target organs like brain, liver , muscle specific micro RNA. Another  method is to alter the  viruses  so as to produce  immune–stimulating chemicals.

Cure for Multiple Myeloma

A clinical trial at the Mayo Clinic suggests that a altered  version of the measles virus could be used to aim at the  cancer cells and put the condition into absolution. Scientist  intravenously administered 10,000 times the typical dosage of measles vaccine to two women, 49- and 65-years-old, who had multiple myeloma, an unusual cancer affecting white blood cells in bone marrow. The virus, that was modified to target cancer cells, eiminated or reduced tumours in the two patients. .In addition to multiple myeloma trial, the modified measles virus is being tested in glioblastoma multiforme (brain cancer) and ovarian cancer.11  The measles virus was genetically modified to contain mammalian NIS gene. On injecting the modified variants of the virus, the tumour cells are bestowed with the capacity to concentrate radioactive iodine i.e. the gene contains information that enables the of iodine from the blood stream to the tumour cells.11  The presence of radioactive iodine within the tumour cells enables easy tracing of the malignant cells with the help of iodine markers.After injecting measles, the patients  suffered from short lived symptoms like fever, low blood pressure and also rapid heart attack.

The over expression of CD46 by the malignant plasma cells(myeloma cells) makes it a target of choice for the measles virus .In short the life cycle of measles virus complements that of myeloma cells. Genetically modified virus gains access to the bone marrow by infecting the RES. The viruses seek and destroy the tumour by multiplying within the tumour cells. The oncolytic effect of the MV-NIS strain can be augmented by administering the β and γ emitter .IMV strains can be retargeted to display a  ligands such asepidermal growth factor receptor vIII, single-chain antibodies against epidermal growth factor receptor, epidermal growth factor receptor vIII, CD38, 30 Her-2/neu, 28 folate receptor α,  31 CD20, 24 and cytokines such as interleukin, targeting receptors highly expressed in tumour cells . An important challenge in the development of MVstrains as cancer therapeutics is preclinical toxicology testing because of the significant limitations of existing animal models as rodents expression of the MV receptors CD46 and SLAMis nil.  Toxicology studies by IV administration of MV-NIS virus was done in cynomolgus monkeys.

Mechanism of Oncolysis

Negative strand RNA paramyxovirus is measles virus. It contains 6 genes that encode 8 proteins, the proteins being

Nucleocapsid (N)

Fusion (F)

Haemagglutinin (H)

Matrix(M)

Large proteins (L)  and small proteins (C and V)

Phospho (P)

The viruses enter the cell by pH independent membrane fusion. The  membrane and receptorfusion takes place which is initiated by F and H proteins respectively. Interaction betweentworeceptor present in the cancer cells namely CD46 and signalling lymphocyte activation system (SLAS) and the H protein takes place. The expression of CD46 helps the tumour cells to escape apoptosis as the cells protect themselves from complement activated lysis. After the process of receptor recognition by the H protein changes of F protein leading to fission and viral entry occur.12 Therefore typical cytophatic effects of measles virus are due to the formation of gaint mononuclear cell aggregates. The production of syncytia can  greatly uplift  the antitumor effect of the virus because, for every infected cell, 50–100 neighbouring cells can  fuse and sanctais formed which is  followed by apoptosis. The derivates of measles virus are tumour specific and has minimal cytophatic effects on non-transformed and normal cells. Measles virus infection  is said to cause profound immunosuppression,  thereby  making  the patients susceptible to secondary infections which inturn accounts  accounts  for high mortality and morbidity. The vaccine strains  and Edmonston strain of measles virus obtained from it is used like a cellular receptor human CD46 but most clinical isolates of measles virus cannot use CD46 as a receptor-5. Transfection with a human SLAM (signalling lymphocyte-activation molecule; also known as CDw150) complementary DNA enables non-susceptible cell lines to combine measles virus and supports measles virus replication and develop cytopathic effects.

The diffusion of SLAM on various cell lines is consistent with their susceptibility to clinical isolates of measles virus. The identification of SLAM as a receptor for measles virus opens the way to a better comprehension of the pathogenesis of measles virus infection, especially the immunosuppression induced by measles virus.13

The current strategies in oncolytic virotherapy are as follows

Overriding  of innate immune response enhances efficacy

Carrier cell technique avoids immune attack

Addressing tumor microenvironment enhances viral spread and efficacy

Oncolytic viruses destroy cancer stem cells

Genetic engineering of oncolytic viruses complements

chemo-and molecular-targeted therapies

Genetic engineering of oncolytic viruses aims cancer signaling pathways

Unique oncolytic virus species are being explored,

Clinical trials

Overriding  innate immune response enhances efficacy

The interaction between  virus-immune system  have been greatly pondered in relation to virotherapy. Innate immune responses to the virus is a prime obstrucle  for long-term gene expression and oncolytic potency. The adoption of immunomodulatory agents in coherence  with oncolytic viruses was first reported in the 1970s . Various studies  demonstrate the  efficacy of  cyclophosphamide to inhibit regulatory T cells  induction, neutralizing antibody induction, macrophages, regulatory T cells  induction and intra-tumoral interferon(IFN)-g production.Though suppression of  immune system enhances the effectiveness  of  the treatment and thereby influencing the overall prognosis to a great extent, it is yet  to be determined ,if  this strategy   would  be beneficial  in  patients with  varying  degree of  previously present   immunosuppression.

Carrier Cell Strategy

By  preventing  the immune  responses one can take exploit the immune system to upgrade antitumor responses. Cytokine-induced killer (CIK) cells destroy tumor cells . After segregating  the CIK cells from mice, these cells were infected with oncolytic vaccines viruses and re –administered  into animals with tumors.  Hence considerably  larger amounts of oncolytic viruses were transported to the tumor. Therefore it was  noted  that both the oncolytic viruses and CIK cells were coherent in tumor killing(12).A  drawback of this approach  is that  it demands harvesting of cells from specific patients, ex vivo nurturing  and re- introduction to the patients and thereby requiring  a substantial amount of laboratory work. Never the less , this approach  holds promise in  expanding the   potency of  the  approach.

Addressing  the Tumor Microenvironment Enhances Viralspread And Efficacy

Tumor microenvironment plays a pivotal  role in limiting  viral spread and enhancing  tumor growth  various approaches have been taken. Coadministration of matrix modifying agents (bacterial collagenase, MMP-1, 8) has demonstrated  to augment   the spread of oncolytic HSV,24,25 although concerns regarding  tumor metastases have to be scrutinized in more preclinical models before translation into clinical trials(13). Tumor hypoxia and its impact on viral replication have also been studied. Inflammation induced by virus infection impacts  the tumor microenvironment. Pretreatment with cyclophosphamide subdued the inflammation and culminated  in decreased tumor vascular permeability.14 Kirn et al.showed that systemically administered vaccinia virus resulted in infection and subsequent destruction of tumor endothelial cells, which led to loss of tumor vascular density.  The efficacy of virotherapy can be limiting when replication-mediated oncolysis is the sole MOA.

Oncolytic Viruses Destroy  Cancer Stem Cells

From the latest explorations in the field of cancer stemcells, it  has  become  evident  that the neoplastic  cell community  not only induce  tumorigenesis, but also contribute  towards resistance to chemo- and radiation therapy.15 As these cell populations  replicatie and self renewl, oncolytic viruses that are constructed  to target cell cycle-dysregulated tumor cells might also possess the potential  to  destroy cancer stem cells. The  mechanism of  action would  incorporate  replication-induced cell annhilation otherwise known as  necrosis and autophagy  that is degradation of intracellular components in lysosomes.

Genetic Engineering

Genetic engineering of oncolytic viruses complementschemo- and molecular-targeted therapie of of the viruses allows functional complementation to chemotherapeutic agents and molecular-targeted therapeutics.15

Ideal Oncolytic Virus Species are Being Explored

As majority of  oncolytic viruses have  exhibited less than optimal efficiency  in clinical trials as solitary agents, there is utmost interest in exploring novel viral species. These studies assess oncolytic activity and  investigate tumor selectivity.

A large number of Clinical Trials have been Carried out

Virotherapy has  an array of features that are unique from  other remedies. Its diverse  innovative MOAs incorporate replication-mediated oncolysis,antitumoral immunity induction, antiangiogenesis, apoptosis and autophage induction. There is no cross resistance with other treatment modalities  and synergistic interaction is exhibited  with other treatment regime. Safety in human has been demonstrated in more than 800 patients.16

Current Trends and Scope of Oncolytic Virotherapy

Although a spectrum  of therapeutic options for  battling neoplasms  inclusive of  surgery, chemotherapy, and local ablative therapies are available, the prognosis for major malignancies remains merger with a median years or months  of  survival. Inspite of marked progress in recent years, most advanced malignancies remain incurable and hence there is an immediate need for the development of novel therapeutics.17 Inspite of exploration of various therapeutic alternates, namely hormonal therapy, immunotherapy, and gene therapy the complete cure for the neoplasms remains a true challenge. The current approach for the treatment  of malignancies is gene therapy, to use viral and non-viral gene therapy systems.18 Gene-based therapeutics has considerable promise as a treat modality. Though gene therapy was originally perceived  as a strategy for treating monogenic diseases, its scope has eventually broadened to incorporate  the in vivo expression of foreign gene products that can produce  tumor cell lysis.19

The efficacy of new generation oncolytic virus is one of the key issues. Increase in anti –tumour activity is being brought about either by incorporating suside genes in the genome or by transiently suppressing the immunity for viral infections. These methods apart from increasing the efficacy also increase the toxicity.19 Higher risks of viral replication are present with immune suppression. This modality of treatment needs a lot of research as there are no proven ways to monitor the in-vivo spread, elimination and for the measurement of viral gene expression and kinetics.20 Cyclophosphamide, a novel strategy is currently being refined  to bypass antimeasles immunity and accelerate systemic delivery in future applications of this technology . One among these notions comprises  the use of cell carriers such as monocytoid cell lines or mesenchymal stem cells, which could protect MV from the immune system, transfer the virus, and efficiently deliver it to tumour cells.21 Intravenously administered viruses are promptly washed off  from the circulation as a result of sequestration by the mononuclear phagocyte system in the liver and spleen.  Prior to clearance, they are opsonised with antibodies, complements, coagulation factors and other serum proteins that enhance their recognition by splenic macrophages and hepatic Kupffer cells. These fragments combine with the  receptors like Fcγ receptors, complement receptor 1 (CR1), CR3 or scavenger receptors on macrophages and endothelial cells, culminating in receptor-mediated phagocytosis and elevated clearance from the circulation.14  An approach to curtail sequestration include chemical alterations of the surface proteins of the viruses by association  of biocompatible polymers, such as polyethylene glycol.

Conclusion

Oncolytic virotheraphy is an emerging field of  cancer biology   that needs  improvement for  implementation as  sole treatment option for cancer.  Logical designing of the viruses based on the knowledge in virology would help to deliver the  virus to the tumour site much effectively with reduced side effects.22 Ex –vivo administration of viruses  prior to  administration to human beings is advised .  Further a critical  biological brink  that has to be exhibited  with all species of oncolytic virus is tumor-selective virus replication, therapeutic transgene expression and biological function.22,23  These developments in the method of treatment help to enhance the prognosis of the patient and also helps to reduce the mortality and morbidity rate due to cancer . The  raising onset , the inadequacy  of effective therapies, and the devastating prognosis of life threatening neoplasm support the immediate  need for new therapeutic agents that are both safe and effective.24,25  These issues if addressed in a timely fashion  and extended  to clinical trials ,virotherapy  will exhibit  great promise as an absolute  treatment manifeston for malignancies with the edge of the potential lack of cross-resistance with standard therapies.

Acknowledgement

The author(s) received no specific funding for this work.

References

  1. Kelly E, Russell S.J. History of oncolytic viruses: genesis to genetic engineering. Molecular Therapy. 2007;1;15(7):651-9.
  2. Gey  G. Tissue culture studies of the proliferative capacity of cervical carcinoma and normal epithelium. Cancer Res. 1952;12:264-5.
  3. Weller T.H, Robbins F.C, Enders J.F. Cultivation of poliomyelitis virus in cultures of human foreskin and embryonic tissues. Proceedings of the Society for Experimental Biology and Medicine. 1949;72(1):153-5
    CrossRef
  4. Cripe T.P, Wang P.Y, Marcato P, Mahller Y.Y,  Lee PW. Targeting cancer-initiating cells with oncolytic viruses. Molecular Therapy. 2009;1;17(8):1677-8.
  5. Cattaneo R, Miest T, Shashkova E.V, Barry M.A. Reprogrammed viruses as cancer therapeutics: targeted, armed and shielded. Nature Reviews Microbiology. 2008; 1;6(6):529-40.
  6. Betancourt D.M. Vesicular Stomatitis Virus is a Malleable Oncolytic Vector for the Treatment of Various Malignancies(Doctoral dissertation, University of Miami).
  7. Singh P.K, Doley J, Kumar G.R, Sahoo A.P, Tiwari A.K. Oncolytic viruses & their specific targeting to tumour cells. The Indian journal of medical research. 2012;136(7):571.
  8. Galanis E. Therapeutic potential of oncolytic measles virus: promises and challenges. Clinical Pharmacology & Therapeutics. 2010;1;88(7):620-5.
  9. Tatsuo H, Ono N, Tanaka K, Yanagi Y. SLAM (CDw150) is a cellular receptor for measles virus. Nature. 2000;406(6798):893.
    CrossRef
  10. Fulci G, Breymann L, Gianni D, Kurozomi K, Rhee S.S, Yu J, Kaur B, Louis DN, Weissleder R, Caligiuri MA, Chiocca EA. Cyclophosphamide enhances glioma virotherapy by inhibiting innate immune responses. Proceedings of the National Academy of Sciences. 2006;22;103(34):12873-8.
  11. Qiao J, Wang H, Kottke T, White C, Twigger K, Diaz RM, Thompson J, Selby P, De Bono J, Melcher A, Pandha H. Cyclophosphamide facilitates antitumor efficacy against subcutaneous tumors following intravenous delivery of reovirus. Clinical Cancer Research. 2008;1;14(1):259-69.
  12. Francica J.R. A study of the Ebola virus glycoprotein: Disruption of host surface protein function and evasion of immune responses (Doctoral dissertation, University of Pennsylvania)
  13. Duke T, Mgone C.S. Measles:not just another viral exanthem. The Lancet. 2003;1;361(9359):763-73.
  14. Lamfers M.L, Fulci G, Gianni D, Tang Y, Kurozumi K, Kaur B, Moeniralm S, Saeki Y, Carette JE, Weissleder R, Vandertop WP. Cyclophosphamide increases transgene expression mediated by an oncolytic adenovirus in glioma-bearing mice monitored by bioluminescence imaging. Molecular Therapy. 2006;1;14(6):779-88.
  15. Liu T.C, Kirn D. Gene therapy progress and prospects cancer: oncolytic viruses. Gene therapy. 2008;15(12):877.
    CrossRef
  16. Fulci G, Dmitrieva N, Gianni D, Fontana E.J, Pan X, Lu Y, Kaufman C.S, Kaur B, Lawler S.E, Lee R.J, Marsh CB. Depletion of peripheral macrophages and brain microglia increases brain tumor titers of oncolytic viruses. Cancer research. 2007;1;67(19):9398-406.
  17. Li H,  Zeng Z,  Fu X, Zhang X. Coadministration of a Herpes simplex Virus-2–based oncolytic virus and cyclophosphamide produces a synergistic antitumor effect and enhances tumor-specific immune responses. Cancer research. 2007;15;67(16):7850-5.
  18. Ungerechts G, Springfeld C, Frenzke ME, Lampe J, Parker WB, Sorscher EJ, Cattaneo R. An immunocompetent murine model for oncolysis with an armed and targeted measles virus. Molecular Therapy. 2007;1;15(7):1991-7.
  19. Di Paolo N.C, Tuve S, Ni S, Hellström KE, Hellström I, Lieber A. Effect of adenovirus-mediated heat shock protein expression and oncolysis in combination with low-dose cyclophosphamide treatment on antitumor immune responses. Cancer research. 2006;15;66(2):960-9.
  20. Liu T.C, Zhang T, Fukuhara H, Kuroda T, Todo T, Canron X, Bikfalvi A, Martuza RL, Kurtz A, Rabkin SD. Dominant-negative fibroblast growth factor receptor expression enhances antitumoral potency of oncolytic herpes simplex virus in neural tumors. Clinical cancer research. 2006;15;12(22):6791-9.
  21. Liu T.C, Zhang T, Fukuhara H, Kuroda T, Todo T, Martuza R.L,  Rabkin S.D, Kurtz A. Oncolytic HSV armed with platelet factor 4, an antiangiogenic agent, shows enhanced efficacy. Molecular Therapy. 2006;1;14(6):789-97
  22. Power A.T, Bell J.C. Cell-based delivery of oncolytic viruses: a new strategic alliance for a biological strike against cancer. Molecular Therapy. 2007;1;15(7):660-5.
  23. Pasquinucci G. Possible effect of measles on leukaemia. The Lancet. 1971;16;297(7690):136.
  24. Bluming A, Ziegler J. Regression of Burkitt’s lymphoma in association with measles infection. The Lancet. 1971;10;298(7715):105-6.
  25. Bell J.C. Taming measles virus to create an effective cancer therapeutic. InMayo Clinic Proceedings.  2014;1;89;7;863-865). Elsevier.
  26. P, Dispenzieri A, Galanis E. Clinical testing of engineered oncolytic measles virus strains in Msaouel the treatment of cancer: an overview. Current opinion in molecular therapeutics. 2009;11(1):43.
  27. Hutzen B, Bid H.K, Houghton P.J, Pierson C.R, Powell K, Bratasz A, Raffel C, Studebaker A.W. Treatment of medulloblastoma with oncolytic measles viruses expressing the angiogenesis inhibitors endostatin and angiostatin. BMC cancer. 2014;14(1):206.
    CrossRef
  28. Cutts F.T, Markowitz L.E. Successes and failures in measles control. Journal of infectious diseases. 1994;1;170(1):32-41.
Share Button
(Visited 692 times, 1 visits today)

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.