Therapeutic Cancer Vaccines; Cancer Treatment Vaccines; Cancer Vaccines; Tumor Vaccines, oncovaccine, tumor antigen vaccines
Cancer treatment vaccines treat cancer by strengthening the body’s natural defenses against the cancer. Vaccines are a type of immunotherapy. Learn how cancer treatment vaccines work against cancer, cancers that are treated with them, and the side effects they may cause.
Hands of a scientist, under a sterile hood, preparing the carcinoembryonic antigen (CEA) vaccinia used to try to prevent cancer.
Image by John Keith (Photographer) National Cancer Institute
How Does It Work?
Vaccine-Based Immunotherapy from Novel Nanoparticle Systems
Image by National Cancer Institute / Victor Segura Ibarra and Rita Serda, Ph.D.
Vaccine-Based Immunotherapy from Novel Nanoparticle Systems
Researchers at the Texas Center for Cancer Nanomedicine (TCCN) are creating particle-based vaccines for cancer therapy. The particles carry molecules that stimulate immune cells and cancer antigens (proteins) that direct the immune response. This scanning electron microscope image shows dendritic cells, pseudo-colored in green, interacting with T cells, pseudo-colored in pink. The dendritic cells internalize the particles, process the antigens, and present peptides to T cells to direct immune responses. This image is part of the Nanotechnology Image Library collection.
Image by National Cancer Institute / Victor Segura Ibarra and Rita Serda, Ph.D.
How Do Cancer Treatment Vaccines Work Against Cancer?
Cancer treatment vaccines are a type of immunotherapy that treats cancer by strengthening the body’s natural defenses against the cancer. Unlike cancer prevention vaccines, cancer treatment vaccines are designed to be used in people who already have cancer—they work against cancer cells, not against something that causes cancer.
The idea behind treatment vaccines is that cancer cells contain substances, called tumor-associated antigens, that are not present in normal cells or, if present, are at lower levels. Treatment vaccines can help the immune system learn to recognize and react to these antigens and destroy cancer cells that contain them.
Cancer treatment vaccines may be made in three main ways.
They can be made from your own tumor cells. This means they are custom-made so that they cause an immune response against features that are unique to your cancer.
They may be made from tumor-associated antigens that are found on cancer cells of many people with a specific type of cancer. Such a vaccine can cause an immune response in any patient whose cancer produces that antigen. This type of vaccine is still experimental.
They may be made from your own dendritic cells, which are a type of immune cell. Dendritic cell vaccines stimulate your immune system to respond to an antigen on tumor cells. One dendritic cell vaccine has been approved, sipuleucel-T, which is used to treat some men with advanced prostate cancer.
A different type of cancer treatment, called oncolytic virus therapy, is sometimes described as a type of cancer treatment vaccine. It uses an oncolytic virus, which is a virus that infects and breaks down cancer cells but does not harm normal cells.
The first FDA-approved oncolytic virus therapy is talimogene laherparepvec (T-VEC, or Imlygic®). It is based on herpes simplex virus type 1. Although this virus can infect both cancer and normal cells, normal cells are able to kill the virus while cancer cells cannot.
T-VEC is injected directly into a tumor. As the virus makes more and more copies of itself, it causes cancer cells to burst and die. The dying cells release new viruses and other substances that can cause an immune response against cancer cells throughout the body.
Source: National Cancer Institute (NCI)
Additional Materials (5)
What is a Cancer Vaccine?
Video by Mayo Clinic/YouTube
Cancer Vaccines: Are we Closer?
Video by Icahn School of Medicine/YouTube
Therapeutic Cancer Vaccines: How They Work | Memorial Sloan Kettering
Video by Memorial Sloan Kettering/YouTube
Cancer Vaccines: Training the Immune System to See Cancer with Dr. Gavin Dunn
Video by Cancer Research Institute/YouTube
Cancer vaccine?: BBC News Review
Video by BBC Learning English/YouTube
4:00
What is a Cancer Vaccine?
Mayo Clinic/YouTube
56:13
Cancer Vaccines: Are we Closer?
Icahn School of Medicine/YouTube
1:03
Therapeutic Cancer Vaccines: How They Work | Memorial Sloan Kettering
Memorial Sloan Kettering/YouTube
39:12
Cancer Vaccines: Training the Immune System to See Cancer with Dr. Gavin Dunn
Cancer Research Institute/YouTube
7:22
Cancer vaccine?: BBC News Review
BBC Learning English/YouTube
tumor-specific antigen
Processing of tumor antigens recognized by CD8+ T cells
Image by UCLLICRBRU
Processing of tumor antigens recognized by CD8+ T cells
Processing of tumor antigens recognized by CD8+ T cells
Image by UCLLICRBRU
tumor-specific antigen
A protein or other molecule that is found only on cancer cells and not on normal cells. Tumor-specific antigens can help the body make an immune response against cancer cells. They may be used as possible targets for targeted therapy or for immunotherapy to help boost the body’s immune system to kill more cancer cells. Tumor-specific antigens may also be used in laboratory tests to help diagnose some types of cancer.
Source: National Cancer Institute (NCI)
Additional Materials (4)
Classes of human tumor antigens recognized by T lymphocytes, with their genetic process
Classes of human tumor antigens recognized by T lymphocytes, with their genetic process
Image by UCLLICRBRU/Wikimedia
The spectrum of target antigens associated with tumor immunity and allo-immunity after allogeneic HSCT.
Host-derived T and B cells can be induced to recognize tumor-associated antigens, whereas donor-derived B and T cells can recognize both tumor-associated antigens and alloantigens.
Image by Wu, C.J., Immunologic targeting of the cancer stem cell (December 15, 2008), StemBook, ed. The Stem Cell Research Community, StemBook, doi/10.3824/stembook.1.21.1, http://www.stembook.org./Wikimedia
Classes of human tumor antigens recognized by T lymphocytes, with their genetic process
UCLLICRBRU/Wikimedia
The spectrum of target antigens associated with tumor immunity and allo-immunity after allogeneic HSCT.
Wu, C.J., Immunologic targeting of the cancer stem cell (December 15, 2008), StemBook, ed. The Stem Cell Research Community, StemBook, doi/10.3824/stembook.1.21.1, http://www.stembook.org./Wikimedia
Image by National Human Genome Research Institute (NHGRI)
Virus
A virus is an infectious agent that occupies a place near the boundary between the living and the nonliving.
Image by National Human Genome Research Institute (NHGRI)
Oncolytic Virus Therapy: Using Tumor-Targeting Viruses to Treat Cancer
For more than a century, doctors have been interested in using viruses to treat cancer, and in recent years a small but growing number of patients have begun to benefit from this approach.
Some viruses tend to infect and kill tumor cells. Known as oncolytic viruses, this group includes viruses found in nature as well as viruses modified in the laboratory to reproduce efficiently in cancer cells without harming healthy cells.
To date, only one oncolytic virus—a genetically modified form of a herpesvirus for treating melanoma—has been approved by the Food and Drug Administration (FDA), though a number of viruses are being evaluated as potential treatments for cancer in clinical trials.
Oncolytic viruses have long been viewed as tools for directly killing cancer cells. But a growing body of research suggests that some oncolytic viruses may work—at least in part—by triggering an immune response in the body against the cancer.
When a virus infects a tumor cell, the virus makes copies of itself until the cell bursts. The dying cancer cell releases materials, such as tumor antigens, that allow the cancer to be recognized, or “seen,” by the immune system.
“Oncolytic viruses are alerting the immune system that something’s wrong,” said Jason Chesney, M.D., Ph.D., director of the University of Louisville’s James Graham Brown Cancer Center. This can lead to an immune response against nearby tumor cells (a local response) or tumor cells in other parts of the body (a systemic response).
For this reason, some researchers consider oncolytic viruses to be a form of immunotherapy—a treatment that harnesses the immune system against cancer. But many in the field would agree that more studies are needed to learn how different oncolytic viruses work against cancer.
A Modern Approach to an Old Idea
Since the late 1800s, doctors have observed that some patients with cancer go into remission, if only temporarily, after a viral infection. Today, several dozen viruses—and a few strains of bacteria—are being studied as potential cancer treatments, according to research presented at an NCI-sponsored conference on using microbes as cancer therapies in 2017.
“Oncolytic virus therapy is of growing interest to researchers for one reason: It’s working,” said Juan Fueyo, M.D., of the University of Texas MD Anderson Cancer Center, who co-developed a type of oncolytic virus being tested in patients with brain tumors.
Although the notion of using viruses in cancer therapy is old, the science only began to move forward in the 1990s with advances in genetic engineering technology, noted Matthias Gromeier, M.D., of the Duke Cancer Institute, who has led clinical trials of a genetically modified form of poliovirus.
“There was another shift—around 2005—as people began to realize that the true value of viruses in cancer therapy is in immunotherapy,” Dr. Gromeier continued. “Today, viruses are firmly established as a potential option to enhance and mediate immunotherapy.”
He added: “These are still early days for oncolytic viruses, but it’s now getting interesting.”
The First FDA-Approved Oncolytic Virus Therapy
The first oncolytic virus to receive FDA approval was a treatment for melanoma known as talimogene laherparepvec (Imlygic), or T-VEC. The treatment, which is injected into tumors, was engineered to produce a protein that stimulates the production of immune cells in the body and to reduce the risk of causing herpes.
In some patients receiving the therapy, tumors that could not be injected have shrunk, suggesting that T-VEC can generate a systemic immune response, noted Howard Kaufman, M.D., of the Rutgers Cancer Institute of New Jersey.
“The oncolytic virus kills tumor cells and causes the release of danger signals, which help to generate an immune response,” explained Dr. Kaufman, who co-led the clinical trial that led to the approval of T-VEC.
Investigating Interactions with the Immune System
At the NCI meeting about using microbes as cancer therapies last year, more than 350 investigators discussed many topics, including the need to better understand how infectious agents interact with tumors and with components of the immune system.
The biological mechanisms used by viruses to kill tumors depend on various factors, including the virus, the target tissue or cell, and which biological pathways are targeted, according to Phillip Daschner of NCI’s Division of Cancer Biology, who helped organize the NCI conference.
Some viruses work primarily by killing tumor cells, whereas others work by directing local or systemic immune responses, he explained. Nonetheless, “there was a consensus at the meeting that even for directly oncolytic therapies, there probably is an important immune component to the response,” he added.
Dr. Kaufman noted that T-VEC, when given alone or in combination with other therapies, generally has been well tolerated by patients in clinical trials.
“We continue to be impressed by the safety profile of these approaches,” he said. “The quality of life for many of these patients is barely affected by these agents.”
Using Viruses to Enhance the Body’s Immune Response
One of the challenges for researchers now is to try to enhance the immune response to the tumor through a variety of strategies, including by combining oncolytic virus therapy and immunotherapy.
The promise of this approach has been demonstrated in two early-phase clinical trials. Patients with melanoma who received T-VEC plus a type of immunotherapy known as a checkpoint inhibitor had higher response rates than those who received a checkpoint inhibitor alone.
In one trial, nearly 200 patients received T-VEC with or without ipilimumab (Yervoy®). The results suggested to the researchers that the combination therapy could induce an immune response. “To me, that’s the big finding of this study,” said Dr. Chesney, who co-led the clinical trial with Dr. Kaufman.
In the second trial, which included 21 patients, T-VEC was combined with pembrolizumab (Keytruda®). The oncolytic virus induced the infiltration of immune cells known as T cells into tumors that had low levels of these cells prior to treatment, the researchers found.
The study suggests that the viral therapy can change the local microenvironment to make an immunologically “cold” tumor—that is, a tumor lacking T cells—into an inflamed, or “hot,” tumor, noted John B.A.G. Haanen, Ph.D., of the Netherlands Cancer Institute in a commentary accompanying the study results.
“The injection [of T-VEC] is like lighting a match—it’s the spark that starts a fire,” said Antoni Ribas, M.D., Ph.D., of the UCLA Jonsson Comprehensive Cancer Center, who led the trial. The therapy was generally well tolerated, he noted, and the most common side effects were fatigue, fever, and chills.
A phase 3 clinical trial involving 600 patients with melanoma who will receive T-VEC with or without pembrolizumab is under way to assess the combination therapy in a large, randomized study.
The same combination—T-VEC plus pembrolizumab—is also being evaluated in a clinical trial for patients with advanced melanoma that has progressed despite treatment with a checkpoint inhibitor such as pembrolizumab or nivolumab (Opdivo®).
This NCI-sponsored trial is testing the idea that injections of T-VEC into accessible melanoma tumors will increase the infiltration of immune cells into these and potentially other tumors, making them susceptible to treatment with pembrolizumab.
A New Way of Delivering Viruses
Most oncolytic virus therapies have been tested in patients with melanoma or brain tumors, and most treatments have been given as injections into tumors. Two new studies highlight efforts to expand the number of cancer types treated with oncolytic virus therapies as well as the methods of delivery.
One of the studies found that an oncolytic virus delivered intravenously could cross the blood–brain barrier and enter brain tumors, killing tumor cells. The treatment uses a type of virus known as a reovirus, which causes mild symptoms of a cold or stomach bug in children.
In the second study, researchers tested the Maraba virus, which was originally isolated from a species of sand fly in Brazil, as a way to sensitize tumors to immunotherapy in a mouse model of triple-negative breast cancer.
In both studies, the researchers found that giving oncolytic virus therapy prior to surgery may alter the body’s immune response and enhance the effects of subsequent treatment with a checkpoint inhibitor.
“Combination immunotherapies like these may be most effective when used early in treatment when tumor burden is less and immune systems are intact,” noted Marie-Claude Bourgeois-Daigneault, Ph.D., of the University of Ottawa, an investigator on the Maraba virus study.
Testing a Modified Form of Poliovirus against Brain Tumors
At the Duke Cancer Institute, Dr. Gromeier and his colleagues have been testing an engineered poliovirus, called PVS-RIPO, in patients with glioblastoma.
When the research began in the mid-1990s, Dr. Gromeier viewed oncolytic viruses primarily as agents for killing cancer cells. His thinking changed, however, as PVS-RIPO was tested in patients, and his team noticed clinical changes associated with immune responses in the patients.
“From the first patients, we observed very clear signs that the virus elicited antitumor immune responses,” Dr. Gromeier recalled. Some patients had swelling in the brain and “profound changes in the tumor that took months to develop, which is consistent with an immune event,” he explained.
Based on the results of the clinical studies, FDA in 2016 granted “breakthrough status” to Duke’s poliovirus therapy, which allows officials at FDA to accelerate the agency’s review of the therapy for approval.
“We are interested in exploring what we can combine with PVS-RIPO,” said Dr. Gromeier. “Everyone seems to agree that no single mode of treatment will do the trick in treating cancer.”
A phase 2 trial testing PVS-RIPO with or without the chemotherapy drug lomustine (Gleostine®) is under way in patients with glioblastoma.
Investigating the Mechanisms of Oncolytic Virus Therapy
To learn more about the mechanisms by which poliovirus therapy attacks tumor cells, the Duke researchers recently conducted experiments in cancer cell lines and in mice.
They found that cancer cells infected with PVS-RIPO released tumor antigens and other material that activated immune cells called dendritic cells and induced an immune response against the cancer cells.
“In the mouse model, we showed that a poliovirus could induce a T-cell response that recognizes the tumor,” said Smita Nair, Ph.D., of the Duke University School of Medicine. The finding provides further support for testing the oncolytic virus in combination with other types of immunotherapies, including checkpoint inhibitors, she added.
The Duke team is planning a clinical trial to test PVS-RIPO in six patients with triple-negative breast cancer. Two weeks before undergoing surgery, the patients will receive injections of the treatment into their tumors and will be followed to determine whether the poliovirus triggers any changes in immune system molecules or in the tumor.
Future Research Questions and Priorities for the Field
As oncolytic viruses are tested in clinical trials, researchers will try to learn which patients are likely to respond. “We need biomarkers to help develop effective combination therapies and to select patients who are most likely to benefit from certain combinations,” said Dr. Nair.
Another challenge for the field will be to use the knowledge gained from the melanoma clinical trials to develop treatments for patients with other types of tumors, Dr. Chesney noted.
For researchers, it will also be important to understand “how much tumor infection and killing is required to treat cancer,” said Dr. Gromeier. “These are critical questions that we need to answer, and we’re working on it.”
Oncolytic Viruses May Also Reveal Insights into Immunotherapy
Research on oncolytic viruses may yield insights into the use of current immunotherapies.
“Viruses are great tools for helping us to understand how the antitumor immune response works,” said Dr. Fueyo of MD Anderson. “What we learn from viruses will help us move the field of immunotherapy forward.”
The evolution in thinking about oncolytic viruses since Dr. Fueyo began working in the field two decades ago represents an important shift that has implications for future research.
“We used to think only about making viruses better—more powerful—at killing tumor cells,” said Dr. Fueyo. “Now we need to find ways to help viruses enhance the immune response.”
Source: National Cancer Institute (NCI)
Additional Materials (1)
Researchers are developing tumor-targeting viruses, like this engineered poliovirus, as potential cancer treatments.
Image by Duke Cancer Institute
Researchers are developing tumor-targeting viruses, like this engineered poliovirus, as potential cancer treatments.
Duke Cancer Institute
Approved Vaccines
Vaccine (1)
Image by John Keith (Photographer)/Wikimedia
Vaccine (1)
Title Vaccine
Description Dr. J. Michael Hamilton preparing the carcinoembryonic antigen (CEA) vaccinia vaccine used to try to prevent cancer. He is diluting the concentrated vaccinia virus into a dose level appropriate for administration to a patient. This vaccinia marks any cancer cells expressing the CEA.
Topics/Categories Treatment -- Biological Therapy
Type Color, Photo
Source National Cancer Institute
Image by John Keith (Photographer)/Wikimedia
Which Cancers Are Treated with Cancer Treatment Vaccines?
Sipuleucel-T is used to treat men with prostate cancer:
that has spread to other parts of the body
who have few or no symptoms
whose cancer does not respond to hormone treatment
T-VEC is used to treat some patients with melanoma that returns after surgery and cannot be removed with more surgery.
Source: National Cancer Institute (NCI)
Sipuleucel-T
Sipuleucel-T Injection
Also called: Provenge®, SipT
Sipuleucel-T (Provenge) is a vaccine used to treat prostate cancer that has spread to other parts of the body. It is used in men who have few or no symptoms and whose cancer has not responded to treatments that lower testosterone levels. Sipuleucel-T is a type of cellular adoptive immunotherapy.
Sipuleucel-T Injection
Also called: Provenge®, SipT
Sipuleucel-T (Provenge) is a vaccine used to treat prostate cancer that has spread to other parts of the body. It is used in men who have few or no symptoms and whose cancer has not responded to treatments that lower testosterone levels. Sipuleucel-T is a type of cellular adoptive immunotherapy.
Provenge is a prescription medicine that is used to treat certain patients with advanced prostate cancer. Provenge is made from a patient’s immune cells that have been treated in the laboratory with GM-CSF (a type of growth factor) and a protein found on prostate cancer cells. Provenge may help the immune system kill prostate cancer cells.
Provenge is approved to treat:
Prostate cancer that has metastasized (spread to other parts of the body). It is used in men who have few or no symptoms and whose cancer is hormone-refractory (does not respond to hormone treatment).
Tell your doctor about all your medical problems, including:
heart problems
lung problems
history of stroke
Tell your doctor about all the medicines you take, including prescription and nonprescription drugs, vitamins, and dietary supplements.
Since Provenge is made from your own immune cells, your cells will be collected approximately 3 days before each scheduled infusion of Provenge. You will need to go to a cell collection center for this collection. The collection is called “leukapheresis” (pronounced loo-kuh-fuh-REE-sis). Your collected cells are sent to a manufacturing center where they are mixed with a protein to make them ready for your infusion.
You will get Provenge in 3 intravenous infusions (put into your veins), about 2 weeks apart. Each infusion takes about 60 minutes. Following each infusion, you will be monitored for at least 30 minutes.
Your doctor will give you a schedule for your cell collection and infusion appointments. It is very important that you arrive on time for your appointments. If you miss an appointment and cannot be infused, your Provenge dose will not be usable. Your doctor will work with you to schedule a new appointment at the cell collection center. You may also get a new infusion appointment.
The most common side effects of Provenge include:
chills
fatigue
fever
back pain
nausea
joint ache
headache
Provenge infusion can cause serious reactions.
Tell your doctor right away if
you have breathing problems, chest pains, racing heart or irregular heartbeats, high or low blood pressure, dizziness, fainting, nausea, or vomiting after getting Provenge. Any of these may be signs of heart or lung problems.
you develop numbness or weakness on one side of the body, decreased vision in one eye or difficulty speaking. Any of these may be signs of a stroke.
you develop symptoms of thrombosis which may include: pain and/or swelling of an arm or leg with warmth over the affected area, discoloration of an arm or leg, unexplained shortness of breath, chest pain that worsens on deep breathing.
you get a fever over 100ºF, or redness or pain at the infusion or collection sites. Any of these may be signs of infection.
Tell your doctor about any side effect that concerns you or does not go away.
These are not all the possible side effects of Provenge treatment. For more information, talk with your doctor.
PROVENGE ® (sipuleucel-T) Suspension for Intravenous Infusion [accessed on Jul 26, 2023]
Sipuleucel-T - NCI. National Cancer Institute. Jun 28, 2010 [accessed on Jul 26, 2023]
Cellular & Gene Therapy Products >> Questions and Answers - Provenge. FDA. [accessed on Mar 16, 2018]
Sipuleucel-T Injection. MedlinePlus/AHFS® Drug Information. [accessed on Oct 29, 2018]
These FAQs provide a summary of the most important information about Sipuleucel-T Injection. If you would like more information or have any questions, talk to your healthcare provider.
T-VEC
Talimogene Laherparepvec Injection
Also called: Imlygic™, T-VEC
Talimogene laherparepvec (Imlygic) is a prescription medicine used to treat certain adults with melanoma that has recurred (come back) after surgery. Talimogene laherparepvec is injected directly into tumors in the skin and lymph nodes. It is a type of oncolytic virus therapy.
Talimogene Laherparepvec Injection
Also called: Imlygic™, T-VEC
Talimogene laherparepvec (Imlygic) is a prescription medicine used to treat certain adults with melanoma that has recurred (come back) after surgery. Talimogene laherparepvec is injected directly into tumors in the skin and lymph nodes. It is a type of oncolytic virus therapy.
Imlygic is a prescription medicine used to treat a type of cancer called melanoma when it is on your skin or in your lymph glands. Imlygic is a weakened form of Herpes Simplex Virus Type 1, which is commonly called the cold sore virus. Your healthcare provider will inject Imlygic directly into your tumor(s).
Imlygic may not help you live longer and may not shrink cancer in your organs (for example, lung or liver).
You should not get Imlygic if you are pregnant or have a weakened immune system (for example, an immune deficiency, blood or bone marrow cancer, steroid use, or HIV/AIDS).
Before getting Imlygic, tell your healthcare provider if you:
Are taking steroids or other medicines that suppress your immune system.
Are taking antiviral medicines to treat or prevent herpes, such as acyclovir.
Have or ever had medical conditions such as: ○ HIV infection or AIDS. ○ Blood or bone marrow cancer. ○ Autoimmune disease. ○ Other medical conditions that can weaken your immune system.
Have close contact with someone who has a weakened immune system or is pregnant.
Are pregnant or plan to become pregnant. ○ Imlygic may harm your unborn baby. ○ You should not become pregnant during treatment with Imlygic. ○ Talk to your healthcare provider about effective birth control methods.
Are breastfeeding or plan to breastfeed.
Tell your healthcare provider about all the medicines you take, including prescription and over-the-counter medicines, vitamins, and herbal supplements. Imlygic may affect the way other medicines work and other medicines may affect how Imlygic works.
Your healthcare provider will inject Imlygic directly into your tumor(s) with a needle and syringe. You will get a second treatment 3 weeks after the first treatment. After that, you will get treatments every 2 weeks for as long as you have tumor(s). You can get treated for 6 months or longer.
Your healthcare provider will decide which tumor(s) to inject and may not inject every one.
It is important to care for the treatment sites properly so that Imlygic does not spread to other people. Your healthcare provider will show you how to do this.
Imlygic virus can spread to other areas of your body or to your close contacts (household members, caregivers, sex partners, or persons sharing the same bed).
Do the following to avoid spreading Imlygic to other areas of your body or to your close contacts:
Avoid direct contact between your treatment sites, dressings, or body fluids and close contacts (for example, use condoms when engaging in sexual activity, avoid kissing close contacts if either has an open mouth sore).
Wear gloves while putting on or changing your dressings.
Keep treatment sites covered with airtight and watertight dressings for at least 1 week after each treatment (or longer if the treatment site is weeping or oozing).
If the dressing comes loose or falls off, replace it right away with a clean dressing.
Place all used dressings and cleaning materials in a sealed plastic bag and throw them away in the garbage.
Do not touch or scratch the treatment sites.
The most common side effects of Imlygic include:
Tiredness
Chills
Fever
Nausea
Flu-like symptoms
Pain at treatment site
Tell your doctor right away if you get any of the signs and symptoms of herpes infection, including but not limited to:
Pain, burning, tingling, or blister formation around the mouth, genitals, or any part of the body, near or far from the injection site
Eye pain, light sensitivity, discharge from the eyes, or blurry vision
Weakness in arms or legs
Extreme drowsiness (feeling sleepy)
Mental confusion
If you think you have a herpes infection, inform your healthcare provider. Life threatening-herpes infection and herpes infection spreading to any part of the body far from the injection site (disseminated herpetic infection) may occur in patients treated with Imlygic. If you have any new or worsening symptoms, call your healthcare provider right away.
These are not all the possible side effects of Imlygic. Your healthcare provider can give you more detailed information. Tell your healthcare provider if you have any side effects that bother you or that do not go away.
DailyMed - IMLYGIC- talimogene laherparepvec injection, suspension [accessed on Dec 07, 2023]
Talimogene Laherparepvec Injection. MedlinePlus/AHFS® Drug Information. [accessed on Oct 29, 2018]
These FAQs provide a summary of the most important information about Talimogene Laherparepvec Injection. If you would like more information or have any questions, talk to your healthcare provider.
Possible Side Effects
Fatigue
Image by TheVisualMD
Fatigue
The state of weariness following a period of exertion, mental or physical, characterized by a decreased capacity for work and reduced efficiency to respond to stimuli. (NCBI/NLM/NIH)
Image by TheVisualMD
What Are the Side Effects of Cancer Treatment Vaccines?
Cancer treatment vaccines can cause side effects, which affect people in different ways. The side effects you may have and how they make you feel will depend on how healthy you are before treatment, your type of cancer, how advanced it is, the type of treatment vaccine you are getting, and the dose.
Doctors and nurses cannot know for sure when or if side effects will occur or how serious they will be. So, it is important to know which signs to look for and what to do if you start to have problems.
Cancer treatment vaccines can cause flu-like symptoms, which include:
fever
chills
weakness
dizziness
nausea or vomiting
muscle or joint aches
fatigue
headache
trouble breathing
low or high blood pressure
You may have a severe allergic reaction.
Sipuleucel-T can cause stroke.
T-VEC can cause tumor lysis syndrome. In this syndrome, the tumor cells die and break apart in the blood. This changes certain chemicals in the blood, which may cause damage to organs like the kidneys, heart, and liver.
Since T-VEC is made from herpesvirus it can sometimes cause a herpesvirus infection that can lead to:
pain, burning, or tingling in a blister around the mouth, genitals, fingers, or ears
eye pain, sensitivity, discharge from the eyes, and blurry vision
weakness in the arms and legs
extreme fatigue and drowsiness
confusion
Source: National Cancer Institute (NCI)
Additional Materials (1)
Fatigue is the Most Common Symptom Experienced by Adults and Children with Cancer
Image by National Cancer Institute
Fatigue is the Most Common Symptom Experienced by Adults and Children with Cancer
National Cancer Institute
MRNA Vaccines
Major delivery methods for mRNA vaccines commonly used delivery methods and carrier molecules for mRNA vaccines are shown
Image by Yang Wang, Ziqi Zhang, Jingwen Luo, Xuejiao Han, Yuquan Wei, and Xiawei Wei/Wikimedia
Major delivery methods for mRNA vaccines commonly used delivery methods and carrier molecules for mRNA vaccines are shown
Major delivery methods for mRNA vaccines commonly used delivery methods and carrier molecules for mRNA vaccines are shown: lipid-based delivery, polyer-based delivery, peptide-based delivery, virus-like replicon particle, cationic nanoemulsion, naked mRNAs and dendritic cell-based delivery.
Image by Yang Wang, Ziqi Zhang, Jingwen Luo, Xuejiao Han, Yuquan Wei, and Xiawei Wei/Wikimedia
Can mRNA Vaccines Help Treat Cancer?
The coronavirus pandemic has thrown a spotlight on messenger RNA (mRNA)—the molecule that carries a cell’s instructions for making proteins. Hundreds of millions of people worldwide have received mRNA vaccines that provide powerful protection against severe COVID-19 caused by infection with SARS-CoV-2.
As stunningly successful as the mRNA COVID-19 vaccines have been, researchers have long hoped to use mRNA vaccines for a very different purpose—to treat cancer. mRNA-based cancer treatment vaccines have been tested in small trials for nearly a decade, with some promising early results.
In fact, scientists at both Pfizer-BioNTech and Moderna drew on their experience developing mRNA cancer vaccines to create their coronavirus vaccines. Now, some investigators believe the success of the mRNA COVID-19 vaccines could help accelerate clinical research on mRNA vaccines to treat cancer.
“There’s a lot of enthusiasm around mRNA right now,” said Patrick Ott, M.D., Ph.D., who directs the Center for Personal Cancer Vaccines at the Dana-Farber Cancer Institute. “The funding and resources that are flowing into mRNA vaccine research will help the cancer vaccine field.”
Dozens of clinical trials are testing mRNA treatment vaccines in people with various types of cancer, including pancreatic cancer, colorectal cancer, and melanoma. Some vaccines are being evaluated in combination with drugs that enhance the body’s immune response to tumors.
But no mRNA cancer vaccine has been approved by the US Food and Drug Administration for use either alone or with other cancer treatments.
“mRNA vaccine technology is extremely promising for infectious diseases and may lead to new kinds of vaccines,” said Elad Sharon, M.D., M.P.H., of NCI's Division of Cancer Treatment and Diagnosis. “For other applications, such as the treatment of cancer, research on mRNA vaccines also appears promising, but these approaches have not yet proven themselves.”
With findings starting to emerge from ongoing clinical trials of mRNA cancer vaccines, researchers could soon learn more about the safety and effectiveness of these treatments, Dr. Sharon added.
How do mRNA vaccines work?
Over the past 30 years, researchers have learned how to engineer stable forms of mRNA and deliver these molecules to the body through vaccines. Once in the body, the mRNA instructs cells that take up the vaccine to produce proteins that may stimulate an immune response against these same proteins when they are present in intact viruses or tumor cells.
Among the cells likely to take up mRNA from a vaccine are dendritic cells, which are the sentinels of the immune system. After taking up and translating the mRNA, dendritic cells present the resulting proteins, or antigens, to immune cells such as T cells, starting the immune response.
“Dendritic cells act as teachers, educating T cells so that they can search for and kill cancer cells or virus-infected cells,” depending on the antigen, said Karine Breckpot, Ph.D., of the Vrije Universiteit Brussel in Belgium, who studies mRNA vaccines.
The mRNA included in the Pfizer-BioNTech and the Moderna coronavirus vaccines instructs cells to produce a version of the “spike” protein that studs the surface of SARS-CoV-2.
The immune system sees the spike protein presented by the dendritic cells as foreign and mobilizes some immune cells to produce antibodies and other immune cells to fight off the apparent infection. Having been exposed to the spike protein free of the virus, the immune system is now prepared, or primed, to react strongly to a subsequent infection with the actual SARS-CoV-2 virus.
Cancer research led to speedy development of mRNA vaccines
When the pandemic struck, mRNA vaccine technology had an unexpected opportunity to demonstrate its promise, said Norbert Pardi, Ph.D., of the University of Pennsylvania Perelman School of Medicine, whose research focuses on mRNA-based vaccines.
“The production of mRNA vaccines today is easy, fast, and can be scaled up as needed,” Dr. Pardi continued. The same manufacturing procedure can be applied to any mRNA sequence, he added.
Historically, the process of developing vaccines has taken 10 to 15 years. But both the Pfizer-BioNTech and the Moderna COVID-19 vaccines—the latter of which was developed in collaboration with NIH—were designed, manufactured, and shown to be safe and effective in people in less than a year.
“To develop an infectious disease vaccine during a pandemic, you need to be fast,” said Lena Kranz, Ph.D., co-director of Cancer Vaccines at BioNTech. “The current pandemic has confirmed our hypothesis that mRNA technology is well suited for fast vaccine development and rapid manufacturing on a global scale.”
The groundwork for the speedy design, manufacturing, and testing of the mRNA COVID-19 vaccines was established through decades of work on cancer vaccines. During this period, immunotherapy, including drugs such as immune checkpoint inhibitors, emerged as a new approach to treating cancer, leading, in some people, to dramatic and long-lasting responses.
“There’s a lot of synergy between research on immunotherapy and mRNA cancer vaccines,” said Robert Meehan, M.D., senior director of clinical development at Moderna. “Vaccines are building on the success of immune checkpoint inhibitors and expanding our knowledge of the underlying biology.”
Modifying and protecting the cargo of mRNA vaccines
Technologies that can deliver mRNA to the body are essential for the success of these vaccines. If an mRNA sequence were injected into the body without some form of protection, the sequence would be recognized by the immune system as a foreign substance and destroyed.
A solution employed by some investigational cancer vaccines is to encase the mRNA in lipid nanoparticles, which are tiny spheres that protect the mRNA molecules. Other delivery vehicles include liposomes, which are also a type of vesicle, or bubble.
“The most advanced mRNA-based vaccine platform uses mRNA encapsulated in lipid
nanoparticles,” said Dr. Pardi. Now that the Pfizer-BioNTech and the Moderna coronavirus vaccine trials have demonstrated the effectiveness of lipid nanoparticles, the technology could certainly be used in future cancer vaccine trials, he added.
Another key feature of the Pfizer-BioNTech and the Moderna coronavirus vaccines is the use of modified forms of mRNA, according to Jordan Meier, Ph.D., of NCI’s Center for Cancer Research, who studies mRNA modifications.
The mRNA in these vaccines incorporates pseudouridine, which is a modification of a naturally occurring nucleoside. Nucleosides are the building blocks of mRNA, and the order of specific nucleosides determines the instructions that mRNA gives to the protein-making machinery in cells.
“The [pseudouridine] modification seems to make the mRNA itself almost invisible to the immune system,” said Dr. Meier. The modification does not alter the function of the mRNA but may enhance the effectiveness of the vaccines, he added.
Cancer researchers have been testing both modified and unmodified forms of mRNA in their investigational treatment vaccines. More research is needed to better understand the relative advantages of each approach for the development of cancer vaccines, Dr. Meier said.
Developing and testing personalized mRNA cancer vaccines
For more than a decade, cancer researchers have been developing a type of treatment known as a personalized cancer vaccine using various technologies, including mRNA and protein fragments, or peptides.
The investigational mRNA vaccines are manufactured for individuals based on the specific molecular features of their tumors. It takes 1 to 2 months to produce a personalized mRNA cancer vaccine after tissue samples have been collected from a patient.
“Speed is especially important for individualized cancer vaccination,” said Mathias Vormehr, Ph.D., codirector of Cancer Vaccines at BioNTech. “A highly individualized vaccine combination must be designed and produced within weeks of taking a tumor biopsy.”
With this approach, researchers try to elicit an immune response against abnormal proteins, or neoantigens, produced by cancer cells. Because these proteins are not found on normal cells, they are promising targets for vaccine-induced immune responses.
“Personalized cancer vaccines may teach the immune system how cancer cells are different from the rest of the body,” said Julie Bauman, M.D., deputy director of the University of Arizona Cancer Center.
Dr. Bauman is co-leading a clinical trial testing a personalized mRNA vaccine in combination with an immune checkpoint inhibitor in patients with advanced head and neck cancer. The study initially included patients with colorectal cancer, but this group did not appear to benefit from the therapy.
For patients with head and neck cancer, however, the early results were positive. Among the first 10 participants, 2 patients had all signs of their tumors disappear following treatment, known as a complete response, and another 5 had their tumors shrink.
“We were surprised to see two complete and enduring responses in our first group of patients with head and neck cancers,” said Dr. Bauman, noting that the study has been expanded to include 40 patients with the disease.
“The number of patients treated is small, but we are cautiously optimistic,” she added. The study is sponsored by Moderna, which makes each personalized vaccine in about 6 weeks.
The manufacturing process starts with the identification of genetic mutations in a patient’s tumor cells that could give rise to neoantigens. Computer algorithms then predict which neoantigens are most likely to bind to receptors on T cells and stimulate an immune response. The vaccine can include genetic sequences for up to 34 different neoantigens.
The promise of personalized immunotherapy with mRNA vaccines is “being able to activate T cells that will specifically recognize individual cancer cells based on their abnormal molecular features,” said Dr. Bauman.
Advancing the science of mRNA cancer vaccines
“A lot of immunotherapies stimulate the immune response in a nonspecific way—that is, not directly against the cancer,” said Dr. Ott. “Personalized cancer vaccines can direct the immune response to exactly where it needs to be.”
Some companies are also investigating mRNA cancer vaccines that are based on collections of a few dozen neoantigens that have been linked with certain types of cancer, including prostate cancer, gastrointestinal cancers, and melanoma.
In addition to clinical trials, fundamental research on mRNA cancer vaccines continues. Some investigators are trying to enhance the responses of immune cells to neoantigens in mRNA vaccines. One study, for example, aims to improve the responses of T cells that become exhausted while attacking tumors.
A challenge for the field is learning how best to identify neoantigens for personalized mRNA cancer vaccines, several researchers said.
“There’s still a lot we need to learn and many questions to answer,” Dr. Ott said. It’s not yet clear, for example, how personalized cancer vaccines should be best combined with other treatments, such as immune checkpoint inhibitors, he added.
As cancer researchers pursue these questions, other investigators will be developing knowledge from the growing number of people around the world who are receiving mRNA coronavirus vaccines.
Insights about the composition of mRNA or the way mRNA is packaged that emerge from studies of viruses could potentially inform work on cancer vaccines, said Dr. Breckpot.
“Unfortunately, it took a pandemic for there to be broad acceptance of mRNA vaccines among the scientific community,” she added. “But the global use of COVID-19 mRNA vaccines has demonstrated the safety of this approach and will open doors for cancer vaccines.”
Source: by Edward Winstead - National Cancer Institute (NCI)
Additional Materials (20)
41541 2020 159 Fig1 HTML
Non-replicating mRNA (NRM) constructs encode the coding sequence (CDS), and are flanked by 5′ and 3′ untranslated regions (UTRs), a 5′-cap structure and a 3′-poly-(A) tail. The self-amplifying mRNA (SAM) construct encodes additional replicase components able to direct intracellular mRNA amplification. (1) NRM and SAM are formulated in this illustration in lipid nanoparticles (LNPs) that encapsulate the mRNA constructs to protect them from degradation and promote cellular uptake. (2) Cellular uptake of the mRNA with its delivery system typically exploits membrane-derived endocytic pathways. (3) Endosomal escape allows release of the mRNA into the cytosol. (4) Cytosol-located NRM constructs are immediately translated by ribosomes to produce the protein of interest, which undergoes subsequent post-translational modification. (5) SAM constructs can also be immediately translated by ribosomes to produce the replicase machinery necessary for self-amplification of the mRNA. (6) Self-amplified mRNA constructs are translated by ribosomes to produce the protein of interest, which undergoes subsequent post-translational modification. (7) The expressed proteins of interest are generated as secreted, trans-membrane, or intracellular protein. (8) The innate and adaptive immune responses detect the protein of interest.
Image by Nicholas A. C. Jackson, Kent E. Kester, Danilo Casimiro, Sanjay Gurunathan, and Frank DeRosa/Wikimedia
mRNA in vitro transcription and innate immunity activation
mRNA in vitro transcription and innate immunity activation. (A) mRNA in vitro transcription. Using DNA with the antigen-encoding sequence as template, mRNA in vitro transcription products contain single-stranded RNA (ssRNA), double-stranded RNA (dsRNA), etc. The ssRNA structure normally includes five-prime cap (5′ cap), five-prime untranslated region (5′ UTR), open reading frame (ORF) region, three-prime untranslated region (3′ UTR), and poly (A) tail structure. (B) RNA translation and antigen presentation. Through endocytosis, mRNAs enter the cytoplasm. Some mRNAs combine with ribosomes of the host cell and translate successfully. Antigen proteins can be degraded to antigenic peptides by proteasome in the cytoplasm and presented to cytotoxic T lymphocytes (CTLs) via major histocompatibility complex (MHC) I pathway. Or, they can be released out of the host cell and taken up by DCs. Then, they are degraded and presented to helper T cells and B cells via MHC-II pathway. B cells can also recognize released antigen proteins. (C) Self-adjuvant effect. Various of pattern recognition receptors (PRRs) can recognize mRNA in vitro transcription product. ssRNA can be recognized by endosomal innate immune receptors (e.g., Toll-like receptor 7 (TLR7), TLR8). dsRNA can be recognized by endosomal innate immune receptors (e.g., TLR3) and cytoplasmic innate immune receptors (e.g., protein kinase RNA-activated (PKR), retinoic acid-inducible gene I protein (RIG-I), melanoma differentiation-associated protein 5 (MDA5), and 2′-5′-oligoadenylate synthase (OAS). Based on those, mRNA products can stimulate the secretion of pro-inflammatory cytokines and type I interferon (IFN), which leads to antigen-presenting cells (APCs) activation and inflammatory reaction. However, they can also activate antiviral enzymes that cause stalled mRNA translation and mRNA degradation.
Image by Shuqin Xu, Kunpeng Yang, Rose Li, and Lu Zhang/Wikimedia
202103 mRNA vaccine
mRNA vaccine
Image by DataBase Center for Life Science (DBCLS)/Wikimedia
Vaccines-08-00776-g001
Key components of in vitro transcribed mRNA that determine the level and duration of expression of the encoded protein. The components that can be modulated are shown in blue, while the effect of modulating these components is shown in green. Abbreviations: 3′ poly-A, three prime polyadenylic acid tail; 5′ cap, five prime cap; PRR, pattern recognition receptor; UTR, untranslated region.
Image by Arthur Esprit, Wout de Mey, Rajendra Bahadur Shahi, Kris Thielemans, Lorenzo Franceschini, and Karine Breckpot/Wikimedia
MRNA therapeutics
Currently, mRNA is mainly delivered to different human tissues through liposomes, lipid nanoparticles(LNPs), polymerase complexes, and cationic peptides, and also has great potential in vaccine development and iPSC cell culture.
A protein called transcription factor is a molecular switch that turns a gene or a cascade of genes on or off. This transcription factor, known TCF1 (for T cell factor-1). is essential for the creation and persistence of disease-fighting antibodies in the bloodstream.
Image by Darryl Leja, NHGRI
Translation Occuring Inside Ribosome (Green) in Process of Protein Synthesis
The process of protein synthesis begins within the nucleus of a cell, where DNA resides. The first step is to reveal the blueprint. In the process called transcription, general transcription factors and RNA polymerase bind to the promoter region of a DNA nucleotide. The DNA helix is first unwound or \"unzipped\" to divulge the instructions for assembling a particular type of protein molecule. The instructions are then copied, or transcribed, to mRNA (messenger RNA), a snake-like strand that will carry the blueprint off for production (blue). When transcription is complete, the mRNA exits the cell nucleus with its duplicate of the blueprint and shuttles off to the protein-making factory, the ribosome (green). Pictured here, the mRNA remains at the ribosome factory and is itself used as the template for protein production. The strand is long and feeds in one side of the ribosome and out the other, advancing like a strip of film through a projector. Production commences as the amino acid \"parts\" arrive at the factory. Amino acid molecules from within the cell are transported to the ribosome by tRNA (transfer RNA, yellow) and dock to the mRNA's assembly line one by one. Each amino acid is adjoined to another by a peptide bond, creating a chain (red). The selection and sequence of amino acids in the chain, as determined by the blueprint, are central to defining which type of protein is being produced.
Image by TheVisualMD
Cancer-busting vaccines are coming: here's how they work
Video by nature video/YouTube
What is a Cancer Vaccine?
Video by Mayo Clinic/YouTube
A Vaccine for Breast Cancer?
Video by Healthcare Triage/YouTube
First Breast Cancer Vaccine Trial Participant Shares Experience
Video by Cleveland Clinic/YouTube
Therapeutic Cancer Vaccines: How They Work | Memorial Sloan Kettering
Video by Memorial Sloan Kettering/YouTube
When Will We Have A Cancer Vaccine | UW Medicine Advancement
Video by UW Medicine/YouTube
How mRNA cancer vaccines work
Video by Science in Motion/YouTube
How mRNA could help fight deadly cancer | DW News
Video by DW News/YouTube
Navigating Cancer Immunotherapy: Exploring the World of mRNA Vaccines
Video by Cancer Research Institute/YouTube
Inside The lab Working To Develop An mRNA Vaccine For Colon Cancer
Video by NBC News/YouTube
MRNA vaccine for pancreatic cancer shows early promise
Video by ABC 7 Chicago/YouTube
Could COVID vaccine technology cure cancer? | DW News
Video by DW News/YouTube
41541 2020 159 Fig1 HTML
Nicholas A. C. Jackson, Kent E. Kester, Danilo Casimiro, Sanjay Gurunathan, and Frank DeRosa/Wikimedia
mRNA in vitro transcription and innate immunity activation
Shuqin Xu, Kunpeng Yang, Rose Li, and Lu Zhang/Wikimedia
202103 mRNA vaccine
DataBase Center for Life Science (DBCLS)/Wikimedia
Vaccines-08-00776-g001
Arthur Esprit, Wout de Mey, Rajendra Bahadur Shahi, Kris Thielemans, Lorenzo Franceschini, and Karine Breckpot/Wikimedia
Translation Occuring Inside Ribosome (Green) in Process of Protein Synthesis
TheVisualMD
4:10
Cancer-busting vaccines are coming: here's how they work
nature video/YouTube
4:00
What is a Cancer Vaccine?
Mayo Clinic/YouTube
4:28
A Vaccine for Breast Cancer?
Healthcare Triage/YouTube
1:41
First Breast Cancer Vaccine Trial Participant Shares Experience
Cleveland Clinic/YouTube
1:03
Therapeutic Cancer Vaccines: How They Work | Memorial Sloan Kettering
Memorial Sloan Kettering/YouTube
2:03
When Will We Have A Cancer Vaccine | UW Medicine Advancement
UW Medicine/YouTube
1:03
How mRNA cancer vaccines work
Science in Motion/YouTube
7:04
How mRNA could help fight deadly cancer | DW News
DW News/YouTube
22:20
Navigating Cancer Immunotherapy: Exploring the World of mRNA Vaccines
Cancer Research Institute/YouTube
3:47
Inside The lab Working To Develop An mRNA Vaccine For Colon Cancer
NBC News/YouTube
0:32
MRNA vaccine for pancreatic cancer shows early promise
ABC 7 Chicago/YouTube
6:35
Could COVID vaccine technology cure cancer? | DW News
DW News/YouTube
MRNA Vaccines Pancreatic cancer
MRNA Vaccine for pancreatic cancer
Image by Sverdrup at English Wikipedia
MRNA Vaccine for pancreatic cancer
Messenger RNA and pancreas
Image by Sverdrup at English Wikipedia
An MRNA Vaccine to Treat Pancreatic Cancer
At a Glance
A personalized mRNA vaccine against pancreatic cancer created a strong anti-tumor immune response in half the participants in a small study.
The vaccine will soon be tested in a larger clinical trial. The approach may also have potential for treating other deadly cancer types.
An experimental vaccine for pancreatic cancer showed progress against the disease. MattL_Images / Shutterstock
Pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic cancer, is one of the deadliest cancer types. Despite modern therapies, only about 12% of people diagnosed with this cancer will be alive five years after treatment.
Immunotherapies—drugs that help the body’s immune system attack tumors—have revolutionized the treatment of many tumor types. But to date, they have proven ineffective in PDAC. Whether pancreatic cancer cells produce neoantigens—proteins that can be effectively targeted by the immune system—hasn’t been clear.
An NIH-funded research team led by Dr. Vinod Balachandran from Memorial Sloan Kettering Cancer Center (MSKCC) have been developing a personalized mRNA cancer-treatment vaccine approach. It is designed to help immune cells recognize specific neoantigens on patients’ pancreatic cancer cells. Results from a small clinical trial of their experimental treatment were published on May 10, 2023, in Nature.
After surgery to remove PDAC, the team sent tumor samples from 19 people to partners at BioNTech, the company that produced one of the COVID-19 mRNA vaccines. BioNTech performed gene sequencing on the tumors to find proteins that might trigger an immune response. They then used that information to create a personalized mRNA vaccine for each patient. Each vaccine targeted up to 20 neoantigens.
Customized vaccines were successfully created for 18 of the 19 study participants. The process, from surgery to delivery of the first dose of the vaccine, took an average of about nine weeks.
All patients received a drug called atezolizumab before vaccination. This drug, called an immune checkpoint inhibitor, prevents cancer cells from suppressing the immune system. The vaccine was then given in nine doses over several months. After the first eight doses, study participants also started standard chemotherapy drugs for PDAC, followed by a ninth booster dose.
Sixteen volunteers stayed healthy enough to receive at least some of the vaccine doses. In half these patients, the vaccines activated powerful immune cells, called T cells, that could recognize the pancreatic cancer specific to the patient. To track the T cells made after vaccination, the research team developed a novel computational strategy with the lab of Dr. Benjamin Greenbaum at MSKCC. Their analysis showed that T cells that recognized the neoantigens were not found in the blood before vaccination. Among the eight patients with strong immune responses, half had T cells target more than one vaccine neoantigen.
By a year and a half after treatment, the cancer had not returned in any of the people who had a strong T cell response to the vaccine. In contrast, among those whose immune systems didn’t respond to the vaccine, the cancer recurred within an average of just over a year. In one patient with a strong response, T cells produced by the vaccine even appeared to eliminate a small tumor that had spread to the liver. These results suggest that the T cells activated by the vaccines kept the pancreatic cancers in check.
“It’s exciting to see that a personalized vaccine could enlist the immune system to fight pancreatic cancer—which urgently needs better treatments,” Balachandran says. “It’s also motivating as we may be able to use such personalized vaccines to treat other deadly cancers.”
More work is needed to understand why half the people did not have a strong immune response to their personalized vaccines. The researchers are currently planning to launch a larger clinical trial of the vaccine.
—by Sharon Reynolds
Source: nih.gov
Additional Materials (6)
pancreas Pre-mRNA
Image by Vossman/Wikimedia
Vaccine
Image by Artem Podrez/Pexels
MRNA therapeutics
Currently, mRNA is mainly delivered to different human tissues through liposomes, lipid nanoparticles(LNPs), polymerase complexes, and cationic peptides, and also has great potential in vaccine development and iPSC cell culture.
Image by Mmc9719/Wikimedia
Pancreatic Cancer
Image by medlineplus.gov
Messenger RNA
The interaction of mRNA in a cell.
Image by Sverdrup at English Wikipedia
Pre-mRNA-1ysv-tubes
A hairpin loop from a pre-mRNA. Notice its bases (light green) and backbone (sky blue). NMR structure of the central region of the human GluR-B R/G pre-mRNA, from the protein data bank ID 1ysv. Taken from w:PDB ids 1ysv model 1 imaged using w:UCSF Chimera.
Send this HealthJournal to your friends or across your social medias.
Vaccines to Treat Cancer
Cancer treatment vaccines treat cancer by strengthening the body’s natural defenses against the cancer. Vaccines are a type of immunotherapy. Learn how cancer treatment vaccines work against cancer, cancers that are treated with them, and the side effects they may cause.