Kanjarpane

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

5

Arjun Kanjarpane
IR-3/09GT
5.31.18
Annotated Source List

Ansel, Ashley, et al. “Monitoring the Efficacy of Oncolytic Viruses via Gene Expression.” ​Frontiers
in Oncology​, vol. 7, Nov. 2017, doi:​10.3389/fonc.2017.00264​.

This review published in ​Frontiers in Oncology ​describes methods by which to monitor


and gauge the efficacy potential of oncolytic viruses. This article primarily mentions gene
expression in tumor cells. This process consists of measuring alterations in gene expression
before and after virotherapy. The magnitude of this change displays the effect of the oncolytic
virus. This article also mentions inserting a ‘transgene’ into the virus, measuring the expression
and interaction with other tumor genes inside the tumor. The researchers found that the
interleukin gene was upregulated ​in vivo ​when placed into murine models using the Vesicular
Stomatitis Virus (VSV). The transgene device, compared to the one previously mentioned
displays greater accuracy when measuring expression.
This article describes a crucial step in using quantitative data to measure oncolytic
virotherapeutic success. While this article features many different charts describing viral types
and the available monitoring devices, it would benefit from having additional descriptive images,
graphics, and other visual tools. This article’s focus is on gene-expression-based kinds of
monitoring and does not explain other types of monitoring such as signaling proteins.

Aref, Sarah, et al. “Measles to the Rescue: A Review of Oncolytic Measles Virus.” ​Viruses​, vol. 8, no.
10, Oct. 2016, pp. 1–16, doi:​10.3390/v8100294​.

This review describes how the Measles Virus has been retargeted to select tumor cells
preferentially. Currently, the primary means by which Measles is attracted to cancer cells is the
overexpression of the CD46 receptor found in tumor cells. Only genetically-modified strains of
the Measles Virus can use the CD46 receptor as a gateway to infect tumors. Such viruses
include the Measles Virus-Edmonston (MV-EDM), MD-CES, and MV-NIS. Wild strains of the
Measles Virus infect cells using receptors to include ‘signaling lymphocyte activation molecule’
(SLAM) and nectin-4 which allow the virus to harm bodily systems. Researchers inserted
tumor-specific ligands to antigens such as carcinoembryonic antigens (CEAs) or the viral
receptors CD20 and CD38, to reprogram the MV-EDM. These antigens/viral receptors are then
selectively found by the virus, which then infiltrates the appropriate cells. Binding the virus to
other known receptors using microRNA targeting sequences are also reprogramming methods.
The article explicitly mentions how the glycoproteins hemagglutinin and fusion proteins
mediate virus attachment and fusion as well as how they can be reprogrammed to mediate viral
entry. In vivo tracking of the treatment is accomplished by using radioactive nickel sulfide
(NIS) dye. To increase immune potency and diminish tumor resistance, researchers also have
disabled immune checkpoint inhibitors in cases with significant malignancies to fight the spread
of cancer. Two of the eight encoding proteins, V and C primarily prevent type-1 interferon
immune responses. This article also mentions how antiviral host immune responses are a
leading cause of oncolytic virotherapeutic failure as the two systems, the virus, and the immune
system, fail to exhibit qualities of synchronization and pose a threat to each other.
The researcher feels this article supplements his research by not only providing in-depth
details of immunology, virus attachment, and infiltration, but also provides these features
specific to the MV. This article also mentions vital parts of the researcher’s studies including the
role of the immune system in MV-Mediated Oncolysis, MV receptors, MV attachment, MV
entry, and MV in vivo tracking. While the article did pose solutions to possible issues regarding
immune-viral synchronization, these solutions are yet to be tested in vivo and remain in an in
vitro state.

Clements, Derek R., et al. “All That Glitters Is Not Gold: The Need to Consider Desirable and
Undesirable Immune Aspects of Oncolytic Virus Therapy.” ​OncoImmunology​, vol. 5, no. 1, June
2015, doi:​10.1080/2162402X.2015.1057674​.

This article, published in ​OncoImmunology, ​presents the various autoimmune responses


from the tumor and oncolytic virus through means of cytokine production, dysregulation of
immune checkpoints, development of tissue barriers, generation of immunosuppressive cells,
myeloid-derived suppressor cells (MDSCs) and alteration of tumor-produced antigens.
Immunosuppression positively affects viral replication, increasing it by a significant factor. This
increase in viral replication also hinders the immune system’s ability to deploy thymus derived T
lymphocytes (T cells) and bone marrow (bursa) derived B lymphocytes (B cells), reducing the
effect of indirect, immune-directed, oncolysis. The article focuses on MDSCs and their ability to
suppress both, innate and adaptive immunity. These cells cloak the tumor and kill T cells by
direct suppression and deplete essential nutrients needed for T cell activation. Two major groups
of MDSCs have been found in tumors, monocytic and granulocytic (MDSC and G-MDSCs),
respectively. The G- variant suppresses T cells through reactive oxygen species production while
the M- variant suppresses T cells with high arginase enzyme expression.
This article was among the best, if not the best article as it directly pertained to
anti-immune, antiviral responses and their correlations. This article lacked graphs, studies,
statistical and other quantitative data, but specifically described specific proteins processes and
methods in detail. This was a great benefit to the researcher. The researcher also benefited from
the language of the text, which meticulously described the actions of each immune suppressor in
an understandable fashion. This enabled correlation with other articles. Lastly, the article
mentioned immune interactions with the tumor and virus and their interrelations.

Donnelly, Oliver G., et al. “Measles Virus Causes Immunogenic Cell Death in Human
Melanoma.” ​Gene Therapy​, vol. 20, no. 1, Jan. 2013, pp. 7–15, doi:​10.1038/gt.2011.205​.

This scholarly study outlines how the Measles Virus (MV) induces tumor cell death in
human melanoma. The measles virus, compared to other viruses can trigger larger and more
significant immune responses in both an active and passive state. Also, the MV virus allows for
an inflammatory process which releases cytokines, such as interferon and High Mobility Group
Box 1 (HMGB1) signaling proteins. The MV-EDM strain used in vivo demonstrated reduced
amounts of dead dendritic cells, which, if in high numbers can lead to a suppressed immune
system. MV-infected cells also released interferon which helps protect normal tissues by evoking
small amounts of antiviral responses as well as having the ability to indirectly lyse tumor cells.
Normally, the immune system is viewed as a mitigator to oncolytic virotherapeutic success as it
terminates viral replication, making it impossible for viruses to successfully attack malignant
cancers. In many clinical studies, the immune system was mediated to aid viral replication.
This study emphasizes the use of the measles virus, similar to other studies, but also
emphasizes the role of the immune system towards viruses in both, a good, and a bad stance. The
researcher utilized the many graphs, charts, and studies to aid in his comprehension regarding the
viruses’ many characteristics and features. This article did not mention other strategies for the
virus to deal with the immune system besides mentioning immunosuppression sourced outside
the tumor. This absence leads the researcher to further questions such as the effects of additional
immune suppression which, for many patients, may not be an option especially if oncolytic
virotherapy is used in a multimodal approach.

Dornan, Mark H., et al. “First-in-Class Small Molecule Potentiators of Cancer Virotherapy.”
Scientific Reports​, vol. 6, May 2016, p. srep26786, doi:​10.1038/srep26786​.

This scientific report describes the challenges with current oncolytic virotherapeutics:
antiviral immune and tumor responses. While most (75%) tumors do not present substantial
antiviral measures, a few do, posing a significant risk to the success of oncolytic
virotherapeutics. To inhibit the production of interferon beta (IFN-B), an antiviral agent, in
tumors, researchers identified a pyrrole-based compound known as
4-dichloro-5-phenyl-6H-furan-2-one. When implanted into a modified vesicular stomatitis virus
(VSV), viral replication in vivo, ex vivo and in vitro was found to increase as much as 1000-fold
in cancer cells. This substance, however, did not destroy tumor-produced antiviral responses, it
merely slowed the activation of such molecules. Concomitantly, the pyrrole exponentially
deteriorated over time. Changes to the pyrrole chemical structure, such as the removable aryl or
hydroxyl group, eliminated antiviral resistance while other changes did not have such an effect.
Since the researcher is studying immune, tumor, and virus synchronization within
oncolytic virotherapy (OV), this source helped considerably due to its analysis of a solution to a
wide-spread problem existing in highly malignant tumors. This article selectively focuses on two
viruses, the Herpes Simplex Virus (HSV), and ​vesicular stomatitis virus (VSV), but does not
mention other OV viruses such as the Nile, Zika, and Measles viruses. Modifying the chemical
structure of the IFN beta-inhibitors could lead to reduced or non-existent adverse effects.
Although this research had a narrow focus, suppressing antiviral responses from tumors (and the
immune system) is a crucial ingredient to enhancing success of in vivo trials of oncolytic
virotherapy.

Hu, Lulu, et al. “Targeting Autophagy for Oncolytic Immunotherapy.” ​Biomedicines​, vol. 5, no. 1,
Jan. 2017, doi:​10.3390/biomedicines5010005​.

This scholarly journal article published in ​Biomedicines ​outlines the contribution of


autophagy from the virus towards the tumor and how it can aid in limiting antiviral responses.
Oncolytic viruses induce apoptosis, necrosis/necroptosis and autophagic cell death which
increase the number of tumor-associated antigens, and damage-associated molecular patterns.
Enhancing oncolytic immunotherapy may involve inducing adaptive immune responses in
response to increased antigen release secondary to increased levels of virus-induced autophagy.
Increased antigen production is crucial to T cell activation, response, and survival towards
tumors. Modulating autophagy through means of two drugs, RD001 and HCQ
(hydroxychloroquine) can induce further anti-tumor immune responses.
This journal article put the researcher in somewhat of a daze; this article focused on
objects which he previously perceived as insignificant, but now, was described as crucial. Unlike
the other articles regarding autophagy, and how viruses increase this to fight off antiviral
responses, this article modulates and inhibits this process to release antigens. This article
however, fails to mention how the virus seems to avoid any attention from the immune system.
This article did however, contain several reference sources including, ​Combinatorial Strategies
for the Induction of Immunogenic Cell Death, ​which the researcher feels, relates to his topic in
greater detail.

Jacobson, Blake A., et al. “Cap-Dependent Translational Control of Oncolytic Measles Virus
Infection in Malignant Mesothelioma.” ​Oncotarget​, vol. 8, no. 38, June 2017, pp. 63096–109,
doi:​10.18632/oncotarget.18656​.

This research paper from the University of Minnesota describes the use of the MV-EDM
virus strain in treatment for malignant mesothelioma. The nectin-4 viral receptor cited by many
other articles is not required for mesothelioma tumor cells, allowing the MV-EDM strain to
infiltrate cells with the CD46 receptor. This receptor was expressed in greater numbers on these
tumor cells in comparison with non-tumor cells. The study focuses on tumor cell translation and
notes two primary actions, the increase and decrease of tumor cell translation on oncolytic
activity. Stimulants, like interferon gamma I (IFG-I), not only increased tumor cell translation
but correspondingly increased oncolytic activity. When tumor translation repressors were
introduced, (4EASO and 4EGI-1 are cited), oncolytic activity seemed to rise along with virus
potency due to its expanded domain and amount of replication.
Regulating tumor translation in oncolytic virotherapeutics may pose benefits regarding
treatment speed, spread, and other safety measures. The ability to modulate tumor cells directly
correlates to treatment safety, especially when using immunological and viral therapeutics.
Increased tumor translation positively correlates with the efficacy of oncolytic virotherapy due to
enhanced marker expression. This article, however, did not address why tumor translation cannot
be stopped entirely.

Meng, Gang, et al. “Mitophagy Promotes Replication of Oncolytic Newcastle Disease Virus by
Blocking Intrinsic Apoptosis in Lung Cancer Cells.” ​Oncotarget​, vol. 5, no. 15, July 2014, pp.
6365–74, doi:​10.18632/oncotarget.2219​.

This scholarly article published in ​Oncotarget ​presents evidence on the use of autophagy
and mitophagy by oncolytic agents, specifically the Newcastle Disease Virus (NDV). Dr. Meng
and his team of researchers report that virus-induced autophagy and autophagic flux were present
in infection of the virus and viral replication increased accordingly, but at the expense of
apoptosis which increased oncolysis. These researchers first presented that the NDV induced
autophagy by displaying autophagosome formation and the presence of SQSTM1, an adaptor
protein used to carry contents to autophagosome-lysosome fusion. These researchers also
investigated the effect of autophagy on apoptotic cell death. When autophagy inhibitor genes
ATG5 were used in the NDV infection, positive apoptotic cells increased at both early and late
stages. With this discovery, the researchers concluded that autophagy and apoptosis both achieve
a common goal, but contradict each other.
While this article did seem to promote the use of mitophagy and autophagy for increased
replication abilities, the article seemed to leave out the larger picture and the goal of oncolytic
viruses: to kill cancer cells. The article helped the researcher to look for and understand further
causes and interconnections between oncolytic processes, specifically autophagy and apoptosis.
The article, however, presented this information without bias and has given the researcher
thoughts about manipulating autophagy to work without hindering apoptotic processes.

Msaouel, Pavlos, et al. “Clinical Testing of Engineered Oncolytic Measles Virus Strains in the
Treatment of Cancer: An Overview.” ​Current Opinion in Molecular Therapeutics​, vol. 11, no. 1,
Feb. 2009, pp. 43–53, ​http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2717625/​.
This author’s manuscript describes the use of oncolytic measles virus strains in cancer
treatment. A weakened strain of the measles virus called Edmonston (MV-EDM) was shown to
infect tumor cells preferentially, while leaving healthy cells unharmed. Overexpression of the
virus receptor CD46 on the tumor surface allowed increased numbers of the virus to infiltrate
tumor cells through membrane fusion. Although the virus also infiltrates cells using the signaling
lymphocyte-activation molecule, (SLAM), this protein is not overexpressed in tumors, rendering
it less effective. Using a replicating vector like the MV-EDM strain, counters tumor
dissemination, increasing treatment efficacy. The MV-EDM strain is explicitly noted as being
very safe, with millions of patients vaccinated. To monitor viral replication and gene expression
in vivo, the MV viruses are engineered to express genes which facilitate imaging (e.g., NIS and
MV-NIS genes). Such genes concentrate radioisotopes inside cells rendering them detectable by
imaging technologies. The use of the MV for oncolytic purposes has been demonstrated against
ovarian, glioblastoma, multiple myeloma cancers, along with many others. During in vitro
testing, the MV-EDM lysed 80-100% of all glioma and ovarian tumor cells when measured
seven days post-infection.
The researcher regards this source as useful. This manuscript while noting the benefits of
measles virus, did not include a detailed discussion of its challenges and shortcomings. This
article did discuss the use of signaling proteins and their use in virotherapy, which the researcher
found to be very helpful. The described clinical studies presented valuable information in mouse
models, setting the stage for clinical trials. For the researcher’s future studies, this article
presented crucial procedures and biological behaviors found in the Measles Virus which can act
with or against other molecular biological processes used to increase efficacy.

Olagnier, David, et al. “Activation of Nrf2 Signaling Augments Vesicular Stomatitis Virus Oncolysis
via Autophagy-Driven Suppression of Antiviral Immunity.” ​Molecular Therapy​, vol. 25, no. 8,
Aug. 2017, pp. 1900–16, doi:​10.1016/j.ymthe.2017.04.022​.

This original, scholarly article published in ​Molecular Therapy ​describes the critical role
of transcription factor Nrf2, which increases viral replication in vesicular stomatitis virus (VSV)
and improves tumor sensitivity to virus infection. More importantly, this article focuses on how
the Nrf2 factor disrupts normal interferon signaling functions, limiting antiviral responses
through autophagy. In murine models, the study found that tumors infected with the antioxidant
compound sulforaphane (SFN) and VSV had a 55.1% more infected rate than cells infected only
with the vesicular virus. This increased infection rate is partly due to SFN’s ability to trigger
autophagy in cancer cells, which deactivate signaling proteins. Reduced antiviral responses
correlated with HO-1 expressions, as well as the inhibition of STAT and STING proteins due to
the presence of SFN.
The researcher hypothesized that this article is among the best; directly correlating to the
benefits of cellular autophagy (or mitophagy) on increased lysed tumor cells. This article also
provided studies, detailed descriptions, and vast reference sources. Despite its benefits, this
article failed to mention how autophagy could be used as a unimodal approach to treat cancer.
The approach taken to present information was immediately focused, as opposed to a broad
analysis which then narrowed.

Pease, Daniel F., and Robert A. Kratzke. “Oncolytic Viral Therapy for Mesothelioma.” ​Frontiers in
Oncology​, edited by Benjamin Gesundheit, vol. 7, no. 179, Aug. 2017, pp. 1–15,
doi:​10.3389/fonc.2017.00179​.

This review covers three main topics; antitumor efficacy, the virus’ ability to
preferentially infect tumor cells and the virus’ ability to deal with immune responses. This article
specifically focused on two viruses, the Herpes and Measles Viruses. The article also displays
the benefits of replicating vectors (over nonreplicating vectors). Replicating viruses allow for
broad treatment areas to treat malignant cancers and in doing so, can be more efficacious due to
its vast quantity. The researchers stated that they had modified the adenovirus to overexpress the
adenovirus death protein (ADP), a protein which kills host cells. In studies, the researchers
modeled the insertion of the HSV thymidine kinase gene which metabolizes ganciclovir into a
toxic byproduct. Infected cells were lysed due to the ganciclovir. Some viruses, such as the
Herpes and Measles viruses, already have a preference towards tumor cells due to an
overexpression of the surface proteins which control a virus’ entry into that cell. For the measles
virus, the protein CD46 is overexpressed and causes a larger amount of the measles virus to enter
the tumor cell. For the herpesvirus, the mediator protein is known as HSV-1.
This article’s use of clinical studies, multiple areas of research placed the researcher in an
intersection allowing the researcher to explore each area enabling further research. Systematic
diagrams, tables, and other visual devices also aided the researcher by providing another means
of information delivery. This article did lack however, on the use of statistical analysis, graphs
and images in clinical studies which subtracted from ease of overall comprehension. Since this
was a broad article, researchers would also benefit from displaying the ongoing issues with the
treatment and possible solutions.

Rajani, Karishma R., and Richard G. Vile. “Harnessing the Power of Onco-Immunotherapy with
Checkpoint Inhibitors.” ​Viruses​, vol. 7, no. 11, Nov. 2015, pp. 5889–901,
doi:​10.3390/v7112914​.

This scholarly study presented in the ​Viruses ​journal specifically describes the use of
anti-checkpoint inhibitors, their benefit to indirect oncolysis and antitumor immune responses.
Tumors evade immune detection by deploying checkpoint proteins which abrogate T cell
communications. To allow T cells infiltration, foreign antibodies block checkpoint proteins
which the tumor expresses; these include CTLA-4, PD-1 and have been approved for treatment.
When tumor cells are lysed, they release damage-associated molecular pattern signals and
tumor-associated antigens. Viruses within tumor cells also provide “pathogen-associated
molecular patterns” known as PAMP. These patterns are recognized by tumor-infiltrating
immune cells which relay signals to antigen presenting cells, APC which release T cells. These T
cells trigger both, an antiviral, and an anti-tumor response. CTLA-4 and PD-1 are both T cell
checkpoint inhibitors and absorb signals presented by the antigen presenting cells prohibiting T
cells from being released. The researchers in this article engineered genes in the measles viruses
which released medication killing the PD-1 and CTLA-4 checkpoint inhibitors.
This report was, concise and directly correlated to the researcher's topic. The researchers
in this article primed the immune system for the anti-tumor response, notably after viral
oncolysis, but failed to mention how they would accomplish this. This article did not mention
means to delay virus-produced immune-stimulating antigens, but rather assumed there was a
preexisting pathway to achieve this goal. The researcher must further his research to find ways to
delay the production of said antigens to ensure viral replication and survival.

Randall, Richard E., and Stephen Goodbourn. “Interferons and Viruses: An Interplay between
Induction, Signalling, Antiviral Responses and Virus Countermeasures.” ​Journal of General
Virology​, vol. 89, no. 1, 2008, pp. 1–47, doi:​10.1099/vir.0.83391-0​.

This scholarly review published in ​Microbiology Society ​presents the increasing role of
interferon in the management of immune-based antiviral responses. Interferons, in the strict
biological sense, are cytokines which are contained in most cells. These cytokines can induce
significant viral resistance upon viral stimulation. Viruses can still replicate due to their ability to
avoid interferon-based antiviral responses. These viruses may avoid these responses by creating
viral dsRNA. The importance of oncolytic viruses are very pronounced; viruses sensitized to
interferon types I, II and III run the risk of reduced effective treatment time and thus, reduced
efficacy. On the other hand, viruses desensitized to interferon along with underlying autoimmune
systems may have prolonged survival times.
This article, while old, presents information about the importance of interferon to the
management and organization of antiviral response: it is one of the primary obstacles facing
oncolytic virotherapy. This 57-page article not only provided a very lengthy in-depth analysis of
the subject, but also provided many charts, graphs and cited various works. This article, due to its
age, presents some incorrect information, as well as some inefficient methods to analyze and
observe the effects of interferon. The researcher can use this information to analyze different
ways in which interferon acts when used in a multimodal fashion and along with other processes
to mitigate antiviral responses.

Shultz, David. “What Does the Measles Actually Do?” ​Sciencemag.Org​, 30 Jan. 2015,
www.sciencemag.org/news/2015/01/what-does-measles-actually-do​.
This article focuses on the specifics of how the Measles Virus functions as well as its
symptoms and effects. The article notes the virus is part of the paramyxovirus family, and comes
from the genus Morbillivirus. The virus, like many others, has no definitive cure but can be
treated with antiviral medications and vaccines derived from the MV-EDM strain. Upon entering
the human body, the virus infects immune cells called macrophages and dendritic cells which
alert the immune system. Following this, infected cells travel to the lymph nodes where the virus
is transmitted to B and T cells which flow through the bloodstream releasing viral particles. Viral
entry into B and T cells is through the CD150 receptor, (along with SLAM and CD46 receptors).
The article notes how the Measles Virus is returning due to people refusing to be vaccinated.
The fast-spreading properties of the Measles Virus may seem to pose an issue for
treatment considerations at first, but due to the use of a relaxed strain, such as the MV-EDM,
widespread replication is far less of an issue. The article does, however, mention the receptors
and replication patterns in which the MV replicates. The author’s mention of increased Measles
cases in the past years has led the researcher to consider other complications which might result
from oncolytic virotherapy. Further research should be conducted to analyze the effects of the
virus on non-vaccinated patients. The researcher also found the article very simple and indirect
which did provide a background to research, but did not facilitate in-depth research.

TEDx Talks. ​Killing Cancer With Viruses: Patrick Lee at TEDxHalifax​. TedXHalifax,
2012, ​www.youtube.com/watch?v=nsoP4SPi2jY​.

This TED​x ​talk with Professor Patrick Lee leads audiences through the process of current
cancer treatments and their drawbacks. Dr. Lee mentions the issue with existing cancer solutions;
they are not efficacious and target all fast-growing cells, and adversely affect hair and nails
which affect the host’s life significantly. Oncolytic viruses, as opposed to conventional cancer
therapies, are self-replicating, self-contained and do not target all fast-growing cells. Dr. Lee also
spoke about how viruses can be modified in vitro, so when in vivo, the viruses can express
signaling or image-traceable proteins. Dr. Lee specifically studied the reovirus, a pathogen of
human descent without major symptoms and its use in OV therapy. While speaking, Dr. Lee
described the process of viral infiltration and infection in which a virus binds to a viral-receptor
and then replicates in the cell until it bursts. Professor Lee also expressed that the treatment
would not involve much human interaction due to, “The chain-reaction [that] leads to all
susceptible cells being killed.”
Similar to other sources, Lee describes the process of viral infiltration while providing an
overview of oncolytic virotherapy. The weight of this source pertains to the presenter, who in his
retired state may have the ability to serve as the researcher’s advisor. While this was just a short
presentation which omitted several key points to successful oncolytic virotherapeutic treatment,
the source did emphasize the use of signaling proteins and the importance of using replicating
viruses or non-replicating virus. The researcher gained information of viral infiltration and
infection and how this information is used in oncolytic viruses to increase efficacy.

Thomas, Maria A., et al. “Immunosuppression Enhances Oncolytic Adenovirus Replication and
Antitumor Efficacy in the Syrian Hamster Model.” ​Molecular Therapy: The Journal of the
American Society of Gene Therapy​, vol. 16, no. 10, Oct. 2008, pp. 1665–73,
doi:​10.1038/mt.2008.162​.

In this author’s manuscript for ​Molecular Therapy, ​researchers presented how


immunosuppressive environments help oncolytic adenovirus replication and increase antitumor
efficacy. Problems currently facing oncolytic virotherapy (OV) stem from the immune response
initiated after virus introduction into the host environment. To create an immunosuppressive
environment, cyclophosphamide (CP) was used due to its effect on the entire body and entire
immune system. After metabolic activation in the liver, which produces
4-hydroxycyclophosphamide, the chemical spreads into cells in which it decomposes into
phosphoramide mustard. The phosphoramide mustard helps in DNA alkylation and apoptosis.
Other uses for CP outside of OV, include organ transplants to prevent rejection and destruction
of foreign body components. When compared to the control group, not given CP or any
adenovirus treatment, cancer volumes increased during the testing time while hamsters only
given virus treatment with an adenovirus saw tumor relapse around weeks 3 and 4. The last
group, which received both, CP and adenovirus treatment saw significant tumor volume decrease
continuing into week 3 and 4 and increased virus titer, which signaled increased viral replication.
This author’s manuscript presented an experiment directly correlating to the researcher's
second control: drug-modulated immunosuppression. This source, however, did lack in
presenting possible drawbacks from using immunosuppressive substances about susceptibility
towards disease and other foreign bodies. Such an immunosuppressive environment ​in vivo ​and
outside of sterile settings could present possibly fatal circumstances for patients, similar to
complications faced by those with HIV infection. However, the manuscript did present a
specific contender (CP) for activating an immunosuppressant environment without other direct
side effects which can help the researcher during future data collection. The levels of the
independent variable also help magnify immune responses as a problem facing oncolytic
virotherapeutics evidenced by the decrease in viral titer in an immunocompetent environment.

Vile, Richard G. “How To Train Your Oncolytic Virus: The Immunological Sequel.” ​Molecular
Therapy​, vol. 22, no. 11, Nov. 2014, pp. 1881–84, doi:​10.1038/mt.2014.188​.

This commentary by Richard G. Vi​l​e represents a rather simple comparison of oncolytic


virotherapy with the movie, “How to Train Your Dragon.” Vile goes into depth writing explicitly
about how T-cell checkpoint inhibitors can boost immune responses in post-therapy conditions.
Checkpoint inhibition is performed either by constructing a negative-strand RNA measles virus
antibody or inserting the antibodies directly into the tumor site. Vile also mentions how the
measles virus is used due to its preferential ability to replicate in tumor cells. Areas of concern
regarding viral and immune synchronization include the effect of the immune system on the
duration of the treatment, viral replication and spread as well as overall oncolysis. Incorporating
additional genes into the virus, such as the Granulocyte-macrophage colony-stimulating factor,
which attracts a group of white blood cells and provides them with, “miracle grow,” can also
help cytotoxicity.
This article places a complex topic, with its numerous subcategories into an
understandable, enjoyable, and relatable experience, breaking the subject matter down into
simplified terms while conveying the end message. The article’s citing of problems in the
oncolytic virotherapeutic field help the researcher further identify key sources to research, such
as implementing non-mediating genes into the virus to mitigate a severe post-therapy immune
response. Such instances are possible shortcomings for the virotherapeutic treatment.
Furthermore, the citing of possible ways to express suppressing T-cells antibodies also leads the
researcher to further, more complex studies. The article did lack sufficient studies and did not
serve as an official report, but due to its simplicity and the possibility of an advisor, the
researcher found this article to be beneficial to his research.

WAN, JIA, et al. “Strategies and Developments of Immunotherapies in Osteosarcoma.” ​Oncology


Letters​, vol. 11, no. 1, Jan. 2016, pp. 511–20, doi:​10.3892/ol.2015.3962​.

This review published in ​Oncology Letters ​presents the use of immunotherapies to treat
osteosarcoma. Immunotherapy included virus-induced tumor oncolysis. Immunomodulation
through T-cell anti-checkpoint inhibitors using PD-1 and PD-IL medicines, vaccine therapy and
upregulation of the immune system are procedures that are noted. Receptor-mediated
endocytosis can occur from adenovirus infection. Once in the cell’s nucleus, cell transcription
begins (either through early-region 1A or early-region 1B). This process allows the viruses’
DNA to be replicated inside the cell eventually causing virus-induced cell lysis. Deletions in the
ELA or ELB genes result in mutated virus strains, which cannot bind to normal cellular proteins.
However, certain proteins expressed on tumors were seen to be virus-bindable proteins. This
feature allows for certain virus strains to only infect tumor cells. Adenovirus ​dl​1520 contains an
E1B deletion which does not express a kD protein. This protein disables the virus’s ability to
replicate and lyse cells featuring the p53 protein, a tumor suppressor found in normal cells to
prevent cancerous mutations.
This article represented, in full feature depth, the process of adapting an adenovirus to
preferentially replicate to tumor cells without having the virus-receptor overexpressed, which
may still infect healthy cells. While this article mentioned several studies and cited the
importance of each, it lacked in discussing the synthesis of oncolytic virotherapy with
immunotherapy to make a comprehensive antitumor treatment. This article is relevant to the
researcher’s effort, as one of the primary areas of concern is the viruses’ response towards the
immune system and disabling the ability to infect healthy cells. This disabling step is safer than
relying on uncontrolled viral receptors and reduces the possibility of unexpected viral replication
in healthy cells. Furthermore, the researcher benefited from noting the specific proteins which
enable in-tumor viral replication which can be used to prevent viral-induced tumor morbidity.

West, Howard (Jack). “Immune Checkpoint Inhibitors.” ​Journal of American Medical Association
Oncology​, vol. 1, no. 1, Apr. 2015, pp. 115–115, doi:​10.1001/jamaoncol.2015.0137​.

This patient page and graphic describes how immune checkpoint inhibitors function and
their associations to other parts of the immune system. Typically, T cells try to attach to tumor
cells, but proteins known as checkpoint inhibitors turn off T cell responses. Disabling immune
checkpoints with certain drugs allow T cells to attack and lyse cancer cells using granule
exocytosis, death receptor systems or phagocytosis (mentioned in ​CTLA-4 and PD-L1
Checkpoint Blockade Enhances Oncolytic Measles Virus Therapy). ​In studies, it was seen that
removing checkpoint inhibiting drugs increased tumor activity. Reintroducing checkpoint
inhibiting drugs resulted in decreased tumor activity. These findings led researchers to a
conclusion: disabling immune checkpoints can aid in cancer treatment and slow cancer
translation.
This organized information page with its numerous graphics gave the researcher a
well-crafted lesson into checkpoint inhibitors, their use in cancer treatment and their possible
side effects. Various other articles have stated how oncolytic viruses deal a “double dose,” first
lysing the cells through viral infiltration and explosion, and then releasing tumor-specific
antigens which T-cells then pick up and use to attack cells. This second benefit from T-cells
relies on the ability to infiltrate tumor cells without mediation which is only possible by using
checkpoint inhibiting features. While the article did provide valuable information, the
information was lacking in depth and unsupported by trials and studies.

Xia, Mao, et al. “Mitophagy Enhances Oncolytic Measles Virus Replication by Mitigating
DDX58/RIG-I-Like Receptor Signaling.” ​Journal of Virology​, vol. 88, no. 9, May 2014, pp.
5152–64, doi:​10.1128/JVI.03851-13​.

This study published in the ​Journal of Virology describes how Oncolytic Viruses are
subject to the modulatory systems which limit their efficacy, the immune system being the
largest one. Researchers report that the MV-EDM virus uses autophagy, or regulated cell death
to dampen immune effects. This process is mediated by the DDX58/RIG-I receptor, known as
RLRs. The use of autophagy in viruses increased viral replication and disabled type I interferons.
The MV-EDM’s use of unexpected SQSTM1/p62-mediated mitophagy decreased mitochondrial
signaling proteins thereby reducing the viral footprint towards the immune system. Modulating
autophagy in future oncolytic therapy proves to be valuable change point to mediate immune
actions towards the virus during treatment.
By using descriptive wording, graphs, and various images, this study pushed the
researcher’s efforts further, presenting problems and solutions, rather than just issues like some
of the previous articles. The researcher has greatly benefitted through this article through the
means of further research and knowledge regarding viral-immune relations. This article did not
fail to mention the various, individual modulating proteins as well as entry-modulating proteins
contained in the virus which helps the researcher to identify other relevant articles. This article’s
shortcomings are contained in the description of possible in vivo effects of autophagy
modulation and the impact it might have on other viral functions. Researchers should analyze
different impacts of the modulations and seek countermeasures for unexpected consequences.
Also, the article focused selectively on the immune response towards the treatment but ignored
possible tumor responses which may alter the results of their data significantly.

You might also like