The month of "June" is identified as Cancer Immunotherapy Research Awareness Month. This year (2023) marks a decade since cancer immunotherapy gained its first acknowledgment as a notable "breakthrough of the year" by science magazine in 2013.
According to studies, more than thirty cancer types are treatable with immunotherapy, and the objective of this awareness month is to expand this figure and provide life-saving treatments to more individuals dealing with cancer.
How is Cancer Targeted by Immunotherapy?
Conventional approaches by a
clinical research organization to combat cancer, such as radiation therapy and
chemotherapy tend to bring about distressing physical and emotional
repercussions. Unfortunately, their efficacy in treating metastatic cancers is
restrained frequently. Furthermore, these treatment methods seldom lead to a
permanent solution as patients may experience relapses or develop resistance to
the treatment.
Immunotherapy presents a promising avenue for eradicating cancer by empowering the patient's immune system to identify and combat cancer cells. By embracing a fresh perspective from several health research institutes, this method has proven to enhance the survival rates of cancers that exhibit unfavorable clinical outcomes through conventional means, such as metastatic melanoma or pancreatic ductal adenocarcinoma.
An additional advantage of
immunotherapies lies in their potential to attain sustained remission, as the
immune system can detect recurring cancer cells, thereby offering long-lasting
and possibly everlasting safeguards against this debilitating disease.
Different Types Of immunotherapies
Presently, there exists a
multitude of immunotherapy options available to combat the spread of cancer.
These include the following:
1. Immune Checkpoint Inhibitors: These interventions obstruct immune
checkpoints that malignancies exploit to elude the body's natural defense
system.
2. Immunomodulators: By stimulating the body's immune response, these
substances empower the immune system to combat cancer effectively. Cytokines,
adjuvants, and agonists are some examples of immunomodulators employed in
cancer treatment.
3. Adoptive Cell Therapy: This approach involves isolating immune
cells from the patient and modifying or expanding a specific cell population to
recognize cancer antigens. Notable adoptive cell therapies encompass CAR-T
therapy, tumor-infiltrating lymphocytes (TIL) therapy, engineered T-cell
receptor therapy, and natural killer (NK) cell therapy.
4. Targeted Antibodies: Utilizing monoclonal antibodies that target
cancer-specific antigens specifically, these therapies prove effective in
combating cancer. Monospecific, bispecific, or cytotoxic drug-conjugated
antibodies are utilized in this regard.
5. Oncolytic Virus Therapy presents a ray of hope in the realm of
cancer treatment as it involves the strategic alteration of a virus to
specifically target and annihilate cancer cells while leaving healthy cells
unharmed. This innovative approach has garnered recognition from the esteemed
US Food and Drug Administration (FDA), granting approval for IMLYGIC, a
modified herpes virus with the remarkable ability to obliterate metastatic
melanoma cells upon direct injection into the tumor.
6. Cancer Vaccines: These vaccines serve the purpose of training the
immune system to recognize existing cancers or prevent cancer caused by viral
infections. FDA-approved vaccines exist for the prevention of cervical and
liver cancer and the treatment of prostate cancer, metastatic melanoma, and
early-stage bladder cancer.
As our understanding of T cells
and various other immune cell behaviors and interactions with cancer cells
expands, the list of immunotherapy types and specific therapeutic options
continues to grow.
Current Trends in Cancer Immunotherapy Research
Checkpoint Inhibitors
Lately, there has been an increased focus on checkpoint inhibitors, which garnered significant attention when Dr. James Allison and Dr. Tasuku Honjo were awarded the prestigious 2018 Nobel Prize. These brilliant minds at health research institutes uncovered the remarkable manner cancer cells exploit CTLA-4 and PD-1 to evade the immune system.
It's worth noting that the mechanism of action for these two molecules differs substantially. While CTLA-4 is a molecule that suppresses the immune response and is typically employed to ensure it remains under control, limiting T-cell priming through competition with the costimulatory molecule CD28, PD-1 operates differently.
In 2011, the FDA approved the
first-ever checkpoint inhibitor, Ipilimumab. It specifically targets and
inhibits CTLA-4. This inhibition enables T cells to respond to tumour antigens.
In contrast, primed T cells express PD-1, which serves as a suppressor of T-cell function when it engages with PD-L1 found on tumour cells and myeloid cells within the tumour microenvironment (TME).
The dynamic duo of pembrolizumab and nivolumab work their magic by obstructing the PD-1 and PD-L1 interaction within the TME and the tumour itself, thus reviving the weary T cells and reinstating their optimal performance.
Highlighting the limited efficacy of checkpoint inhibitors in cancer treatment, researchers and medical professionals have worked to identify methods for increasing effectiveness and safety.
As a case in point, the FDA
approved the combination of PD-1 inhibitor nivolumab with CTLA-4 inhibitor
ipilimumab to treat metastatic non-small cell lung cancer (NSCLC). Recently,
the agency has also allowed the use of relatlimab - an inhibitor of the immune
checkpoint LAG3 - together with nivolumab for treating unresectable or
metastatic melanoma.
Targeted Antibody Immunotherapy
Targeted antibodies possess the
ability to obstruct the function of specific molecules, trigger programmed cell
death, or regulate signalling pathways. As an illustration, when the monoclonal
antibody known as Herceptin latches onto excessively expressed HER2 receptors
found in breast and stomach cancers, it mitigates the activity of downstream signalling
pathways, thereby instigating apoptosis and halting the growth of cells. Since
approximately 25% of breast cancers exhibit HER2 positivity, Herceptin serves
as a formidable weapon in the existing arsenal of cancer therapies. Although
Herceptin has obtained approval since 1998, researchers persist in exploring
avenues to enhance its efficacy or reduce its toxicity by investigating
alternative epitopes or modifying the antibody to enhance its affinity.
Furthermore, the antibody can be
joined with a chemotherapy payload, creating an antibody-drug conjugate that
selectively targets the cytotoxic molecule in the tumour. Lastly, bispecific
antibodies represent an innovative approach that merges the variable regions of
two distinct antibodies into a single molecule, enabling it to recognize
epitopes from, for instance, both a cancer cell and a T cell. By doing so, the
bispecific antibody brings the T cell into close proximity to the tumour,
resulting in T cell activation, proliferation, and T cell-mediated elimination
of cancer cells. The first bispecific antibody, blinatumomab, received FDA
approval in 2014 for the treatment of leukaemia patients.
Immunotherapies, particularly
utilizing IgG and T cells to help fortify the immune system, have produced
promising outcomes. However, research into more immune cells and
immunoglobulins is being conducted. One of the possibilities being studied entails
immunoglobulin E (IgE) to fight chondroitin sulfate proteoglycan 4 (CSPG4),
which can be present in up to 70% of melanomas. IgE is activated in response to
allergens such as pollen, and it has certain advantages over IgG therapies.
These include its increased affinity for FceRI receptors, lack of inhibitory
receptors for IgE, capacity to bring about a response with various effector
cells in comparison to IgG, low level of existence in the blood with little
opposition for receptor binding, and its capability to induce
antibody-dependent cell-mediated cytotoxicity (ADCC) without having to depend
on complement-mediated cytotoxicity (CDC).
Adoptive Cell Therapy
Individualized medicine has
several faces, and one of them is adoptive cell therapy. In this technique,
cells taken from the patient or donor are modified so that they become
proficient in detecting and destroying tumours - referred to as 'living drugs'.
Notable methods of adoptive cell therapy include tumour-infiltrating
lymphocytes (TILs) and chimeric antigen receptor (CAR) T-cell therapy. After changing
the cells, they are injected back into the individual.
Extracting TILs from tumour
biopsies that can identify multiple cancer antigens, then multiplying their
numbers and administering them to the patient, has demonstrated immense
potential as a viable and secure combination with chemotherapy and IL-2, in
treating melanoma and other solid tumours. Nevertheless, the presence of
signals in the tumour microenvironment typically dampens the activity of these
blood cells.
Due to the difficulty in cultivating
enough TILs and the reality that not all patients have months to wait,
alternative approaches are being investigated. For instance, combining TILs
with checkpoint inhibitors and the development of an engineered TIL with the
FDA's approval, which eliminates the need for IL-2, are among the possible
solutions to expand the reach of this promising therapy.
CAR-T cell therapy has
established itself as a preeminent form of adoptive cell therapy due to its
propensity for potent response rates and long-lasting remission when battling
aggressive leukemias and lymphomas. This procedure entails harvesting and
engineering immune cells from the patient, enabling them to recognize tumour
antigens, thereby inducing tumour cell destruction. The United States Food and
Drug Administration has approved six CAR-T treatments that target acute
lymphoblastic leukaemia in children, B-cell lymphoma, follicular lymphoma,
mantle cell lymphoma, and multiple myeloma in adults.
Despite the potential of CAR-T
therapies, progression towards treating solid tumours is still lagging. These
roadblocks include difficulty recognizing tumour-specific antigens, the TME's
ability to stymie immunotherapy, and the attendant toxicities associated with the
CAR-T cells. In response to these limitations, significant efforts are being
made to circumvent the immunosuppressive power of the TME, for example by
combining CAR-T cell treatments with checkpoint inhibitor drugs, or by
developing CARs capable of disregarding immunosuppressive cues.
The CAR-T process requires the
isolation of T cells, engineering to produce CAR, and validating surface
expression. However, detection reagents with the correct specificity can be
hard to acquire. An alternative method is to validate the CAR-T independently
of antigens using antibodies that target the linker sequence on the scFv
between the variable heavy and light domains.
Cancer Vaccines
Driven by the immediate necessity of a COVID-19 vaccine, advances made by clinical research organizations, in tandem with an improved grasp of the immune system, have now been used to craft cancer vaccines. An initially encouraging therapeutic plan that was mostly disregarded during the 2010s, vaccines that stimulate an immune response to battle castration-resistant prostate cancer and dangerous non-muscle-invasive bladder cancer (NMIBC) have been accepted by the Food and Drug Administration.
With the invention of nucleic
acid vaccines, difficulties related to manufacturing time and administering
live-attenuated vaccines to immunocompromised individuals have been addressed
by health research institutes. Notably, the human papillomavirus (HPV) vaccine
has been developed to protect against cervical cancers and hepatitis B virus
vaccines for liver cancer. Unfortunately, preventative vaccines for non-viral
cancers are not currently available in the clinic. However, researchers are
studying whether improved diagnostic and screening procedures can be coupled
with therapeutic vaccines to stimulate anti-cancer immunity before cancer
progresses to a more advanced stage.
Oncology Research – A Promising Future
Immunotherapy has been a ray of hope for cancer patients, thus providing them some of the most novel treatments. However, it is not a panacea. Joining forces with tried-and-true strategies such as surgery, chemotherapy, and radiotherapy can help medical experts craft tailored treatments for every patient.
Constant novel developments in
science are being discovered. As a recent example, researchers have been
exploring the impact of gut microbiome tuning on patient response to
treatments. An experiment with mice found that microbiota sourced from cancer
patients had an influence on PD-L2 and RGMb protein expression and improved the
responsiveness to anti-PD-1 immunotherapy. Conversely, a broad spectrum of
antibiotics reduced the same response. These results hint at the potential of
improving patient responses to immunotherapy through manipulation of the gut
microbiome.
Thanks to immunotherapies, cancer treatments have been revolutionized, and physicians have an extensive range of options to select from.
As we continue to gain an
understanding of the immune system from studies conducted by clinical research
organizations and design novel approaches to manipulate regulatory systems, the
array of solutions will increase, inching us closer to a future in which we can
ward off and even heal metastatic cancer.
About I3T
In January 2020, Johns Hopkins
University, Baltimore, Maryland, in the United States became a collaborating
partner of the International Institute of Innovation & Technology Kolkata,
India (I3TK). It has a 156,000 square feet state-of-the-art building. It was developed
with a specific goal to research and educate public health-related subjects. As
such, I3T has an Advanced Diagnostic Centre focusing on the exploration of
methods for cancer prevention, detection, and improvement of patient quality of
life through clinical trials.
Comments
Post a Comment