TMM Logo

Tiruvalla Medical Mission (TMM) - Oncology (Cancer) & Nuclear Medicine

This site has been developed for Cancer Patients & their Relatives under the 'Right of information for Patients'.

You may translate this page by clicking on the 'Select Language' - Top Left.

Recommended Products
msm1
About this item 500 Grams, MSM (Methylsulfonylmethane) Powder Guaranteed 100% Pure. No Fillers or Additives. Lab Tested and Verified. Import Origin: USA. Improves flexibility, Detoxifies the body, Strengthens hair and nails, Accelerates energy
In Stock!
Magnesium Oil
Very good for Muscle Cramps. Massage cream on feet, legs, body post sports workout. Lavender sleeping oil helps relax men & women Lavender Aromatherapy is a natural sleep supplement increases sleeping quality, better sleep support
Target Therapy
CANCER - ALL YOU NEED TO KNOW
1. Accurate Diagnosis  >  5. Treatment of Cancer  >  f. Immunotherapy (Target Therapy)

CAR-T therapy

What is Immunotherapy and CAR-T Therapy?

 

Immunotherapy is a type of cancer treatment that uses the body’s immune system to fight cancer. It works by either stimulating the immune system to recognize and attack cancer cells or by providing it with components, such as immune system proteins, to enhance its ability to combat cancer.

 

How Immunotherapy Works in Cancer Treatment

 

1.Boosts Immune Response: Enhances the natural ability of the immune system to detect and destroy cancer cells.

2.Overcomes Immune Evasion: Cancer cells often develop mechanisms to avoid immune detection. Immunotherapy blocks these mechanisms, exposing cancer cells to the immune system.

3.Targets Specific Pathways: Many immunotherapy drugs are designed to target specific molecules involved in immune suppression or cancer growth.

 

Types of Immunotherapy Used in Cancer

 

1.Immune Checkpoint Inhibitors: These drugs block immune checkpoint proteins (e.g., PD-1, PD-L1, CTLA-4) that cancer cells use to suppress the immune system.

Examples:

  • Pembrolizumab (Keytruda) and Nivolumab (Opdivo): Used for melanoma, lung cancer, and others.
  • Atezolizumab (Tecentriq): Used for urothelial carcinoma and non-small cell lung cancer.

2.CAR T-Cell Therapy: Involves engineering a patient’s T-cells in a lab to recognize and destroy cancer cells, then reinfusing them into the patient.

Example:

  • Axicabtagene Ciloleucel (Yescarta): Used for large B-cell lymphoma.

3.Cancer Vaccines: Stimulates the immune system to recognize and attack specific cancer-associated antigens.

Example: Sipuleucel-T (Provenge): Used for metastatic prostate cancer.

4.Cytokine Therapy: Uses proteins like interleukins or interferons to boost the immune system.

Examples:

  • Aldesleukin (IL-2): Used for metastatic renal cell carcinoma and melanoma.
  • Interferon-alpha: Used for some types of leukemia and melanoma.

5.Oncolytic Virus Therapy: Uses genetically modified viruses to infect and kill cancer cells, simultaneously stimulating an immune response.

Example: Talimogene laherparepvec (T-VEC, Imlygic): Used for melanoma.

6.Monoclonal Antibodies: Designed to target specific antigens on cancer cells.

Examples:

  • Rituximab (Rituxan): Used for non-Hodgkin lymphoma and chronic lymphocytic leukemia.
  • Trastuzumab (Herceptin): Used for HER2-positive breast cancer.

 

Examples of Immunotherapy Drugs and Their Applications

 

Drug Type Cancer Types Treated

  • Pembrolizumab (Keytruda) Immune Checkpoint Inhibitor Melanoma, NSCLC, head and neck cancers
  • Nivolumab (Opdivo) Immune Checkpoint Inhibitor Melanoma, renal cell carcinoma, Hodgkin lymphoma
  • Trastuzumab (Herceptin) Monoclonal Antibody HER2-positive breast and gastric cancers
  • Sipuleucel-T (Provenge) Cancer Vaccine Prostate cancer
  • Axicabtagene Ciloleucel CAR T-Cell Therapy Large B-cell lymphoma
  • Atezolizumab (Tecentriq) Immune Checkpoint Inhibitor Urothelial and lung cancers
  • Aldesleukin (IL-2) Cytokine Therapy Renal cell carcinoma, melanoma

 

Benefits of Immunotherapy

 

Specificity: Targets cancer cells without affecting normal cells as much as conventional chemotherapy.

Long-Term Response: Some patients achieve durable remissions even in advanced cancers.

Fewer Side Effects: Compared to chemotherapy, though immune-related side effects like inflammation can occur.

 

Limitations and Challenges

 

Not Universal: Not all patients respond; effectiveness can depend on cancer type and individual biology.

Side Effects: Immune overactivation can cause autoimmune-like symptoms, such as colitis or pneumonitis.

Cost: Immunotherapy drugs are expensive, limiting accessibility in some regions.

--------------

CAR-T (Chimeric Antigen Receptor T-cell) Therapy Protocol

 

Overview of CAR-T Therapy

 

CAR-T therapy is a form of adoptive cell transfer (ACT) that uses genetically engineered T-cells to target and destroy cancer cells. It is primarily used for hematologic cancers like B-cell lymphomas, acute lymphoblastic leukemia (ALL), and multiple myeloma.

 

Steps in the CAR-T Therapy Protocol

 

1.Patient Selection:

  • Suitable for patients with relapsed or refractory cancers.
  • Pre-treatment evaluation includes a thorough medical history, performance status, organ function tests, and imaging.

2.Leukapheresis:

  • •T-cells are collected from the patient’s blood through a process called leukapheresis.
  • •The procedure takes several hours and separates T-cells while returning other blood components to the patient.

3.T-Cell Engineering:

•The collected T-cells are sent to a laboratory or manufacturing facility.

•T-cells are genetically engineered to express a chimeric antigen receptor (CAR) that can recognize a specific antigen on cancer cells (e.g., CD19 in B-cell malignancies).

•This involves introducing a CAR gene into T-cells using viral vectors.

4.Cell Expansion:

•The modified T-cells are cultured and expanded to reach the required therapeutic dose.

•Quality checks ensure the cells are functional and meet safety criteria.

5.Preconditioning Chemotherapy:

•Patients receive lymphodepleting chemotherapy (e.g., cyclophosphamide and fludarabine) a few days before CAR-T infusion.

•This reduces the patient’s existing immune cells, creating space for CAR-T cells to proliferate and function effectively.

6.CAR-T Cell Infusion:

•The engineered CAR-T cells are infused back into the patient through an IV line.

•The process resembles a blood transfusion and is typically completed in 30-90 minutes.

7.Monitoring and Management:

•Patients are closely monitored for side effects, especially during the first few weeks.

•Common side effects include:

Cytokine Release Syndrome (CRS): Fever, hypotension, and organ dysfunction caused by massive cytokine release.

Neurotoxicity (ICANS): Confusion, seizures, or encephalopathy.

•Management includes supportive care and drugs like tocilizumab (for CRS) and corticosteroids (for neurotoxicity).

8.Follow-Up:

•Regular follow-up visits include physical exams, blood tests, and imaging to assess response and detect relapse.

 

FDA-Approved CAR-T Therapies and Their Indications

 

Therapy Name Target Antigen Indications

Axicabtagene Ciloleucel (Yescarta) CD19 Large B-cell lymphoma

Tisagenlecleucel (Kymriah) CD19 Pediatric and young adult B-cell ALL, large B-cell lymphoma

Lisocabtagene Maraleucel (Breyanzi) CD19 Relapsed or refractory large B-cell lymphoma

Idecabtagene Vicleucel (Abecma) BCMA Relapsed or refractory multiple myeloma

Ciltacabtagene Autoleucel (Carvykti) BCMA Relapsed or refractory multiple myeloma

 

Benefits of CAR-T Therapy

 

1.High Efficacy:

•Offers durable remissions in patients with otherwise incurable cancers.

2.Personalized Medicine:

•Tailored to individual patients based on their specific cancer and immune profile.

3.Expanding Applications:

•Research is ongoing to extend CAR-T therapy to solid tumors, though challenges remain.

 

Challenges and Limitations

 

1.High Cost: CAR-T therapy is expensive due to its complex manufacturing process.

2.Limited Availability: Not widely accessible in all countries or hospitals.

3.Side Effects: CRS and neurotoxicity require specialized management, often in an ICU setting.

---------------

CAR-T Implementation for Diffuse Large B-Cell Lymphoma (DLBCL)

 

Case Study Example: Relapsed/Refractory DLBCL

A 56-year-old male with relapsed diffuse large B-cell lymphoma (DLBCL) after initial treatment with R-CHOP (Rituximab + Cyclophosphamide, Doxorubicin, Vincristine, Prednisone) underwent CAR-T therapy with Axicabtagene Ciloleucel (Yescarta).

 

Treatment Steps

 

1. Patient Selection

 

Eligibility: The patient failed two prior lines of chemotherapy, including salvage therapy.

Tests Conducted:

•PET-CT: Showed persistent disease in mediastinal and retroperitoneal lymph nodes.

•Bloodwork: Confirmed adequate organ function (kidney, liver, cardiac).

•ECOG Performance Status: 1 (able to perform daily activities with minimal limitations).

 

2. Leukapheresis

 

•T-cells collected from the patient’s peripheral blood at a specialized facility.

•The process lasted approximately 4-6 hours.

•Cells were sent to a manufacturing lab for genetic modification.

 

3. T-Cell Modification and Expansion

 

•The patient’s T-cells were genetically engineered to express CAR targeting the CD19 antigen, which is highly expressed on DLBCL cells.

•T-cells were cultured and expanded to achieve a therapeutic dose.

•This process took 2-3 weeks.

 

4. Lymphodepleting Chemotherapy

 

•Administered 3 days before CAR-T infusion:

Cyclophosphamide: 500 mg/m² IV daily for 3 days.

Fludarabine: 30 mg/m² IV daily for 3 days.

•Purpose: Reduced the patient’s normal immune cells to allow CAR-T cells to engraft and proliferate effectively.

 

5. CAR-T Cell Infusion

 

•On Day 0, CAR-T cells (target dose of 2 x 10⁶ cells/kg) were infused via IV over 30 minutes.

•The infusion was uneventful, and the patient was monitored for immediate reactions.

 

6. Monitoring and Side Effect Management

 

•The patient was hospitalized for close monitoring during the first 7 days post-infusion.

Cytokine Release Syndrome (CRS):

•Day 3 post-infusion: High fever (102°F), hypotension.

•Managed with Tocilizumab (IL-6 receptor antagonist) and supportive care (IV fluids, antipyretics).

Neurotoxicity (ICANS):

•Day 6 post-infusion: Mild confusion, resolved with low-dose corticosteroids.

•Bloodwork: Monitored for cytokine levels, liver, and kidney function.

 

7. Follow-Up

 

First PET-CT Scan: 30 days post-infusion.

•Result: Significant reduction in tumor size, achieving a partial response.

90-Day Follow-Up:

•PET-CT confirmed a complete metabolic response.

•Regular follow-up every 3 months for the first year and every 6 months thereafter.

 

Outcome

 

•At 12 months post-CAR-T therapy, the patient remained in remission, with no evidence of disease on imaging.

•Quality of life improved, and the patient returned to work part-time.

 

Key Takeaways

 

1.Durable Remission Potential:

•CAR-T therapy provided long-term remission in a patient who had exhausted standard treatment options.

2.Individualized Care:

•Close monitoring and prompt management of CRS and neurotoxicity were critical for success.

3.Follow-Up Importance:

•Regular imaging and blood tests ensured early detection of potential relapse or side effects.

--------------------

Next-Generation CAR-T Therapy

 

Next-generation CAR-T cell therapies are advancing beyond current FDA-approved protocols to address limitations such as relapse, resistance, and restricted efficacy in solid tumors. These innovations aim to improve safety, efficacy, durability, and applicability of CAR-T therapy.

 

Key Advancements in CAR-T Therapy

 

1. Dual-Targeting CAR-T Cells

 

Concept: These CAR-T cells target two antigens simultaneously to prevent tumor escape mechanisms.

Example: Targeting BCMA and CD38 in multiple myeloma or HER2 and EGFR in solid tumors.

Benefit: Reduces the risk of antigen loss and improves efficacy.

 

2. Armored CAR-T Cells

 

Mechanism: These CAR-T cells are engineered to secrete cytokines (e.g., IL-12) or other immune-modulating agents to enhance their activity within the tumor microenvironment (TME).

Benefit: Overcomes immunosuppression in solid tumors and improves CAR-T persistence.

 

3. Universal or Off-the-Shelf CAR-T Cells

 

Concept: Instead of using patient-derived T-cells, universal CAR-T cells are derived from healthy donors and gene-edited to prevent rejection.

Technologies: CRISPR or TALEN editing to knock out genes like TCR or MHC.

Benefit: Reduces manufacturing time and cost, making therapy more accessible.

 

4. Switchable CAR-T Cells

 

Mechanism: These cells include a “safety switch” or a “dual-activation” mechanism where CAR-T cells are activated only in the presence of a specific drug or signaling molecule.

Benefit: Improves safety by allowing clinicians to control CAR-T activity, reducing the risk of severe side effects like cytokine release syndrome (CRS).

 

5. Universal CARs

 

Concept: A universal CAR platform uses adaptable receptors that can bind to multiple tumor antigens through small molecule adaptors.

Benefit: Broadens the range of targetable tumors and improves flexibility.

 

6. CAR-T Cells for Solid Tumors

 

Challenge: Solid tumors have an immunosuppressive TME that limits CAR-T infiltration and function.

Advancements:

•Engineering CAR-T cells with enzymes that degrade the extracellular matrix (e.g., heparanase).

•Equipping CAR-T cells with chemokine receptors to enhance homing to tumors.

•Combining CAR-T therapy with checkpoint inhibitors (e.g., anti-PD-1 antibodies).

 

7. Persistence-Enhanced CAR-T Cells

 

Approach: Incorporation of co-stimulatory domains like 4-1BB and CD28 or adding cytokine support (e.g., IL-7, IL-15).

Benefit: Improves the long-term persistence and activity of CAR-T cells.

 

8. Gamma Delta (γδ) CAR-T Cells

 

Innovation: Use γδ T-cells, which possess inherent anti-tumor properties and are less likely to cause graft-versus-host disease (GVHD).

Benefit: Broadens applicability for allogeneic (donor-derived) CAR-T therapies.

 

Emerging CAR-T Cell Therapies

 

 
Therapy Name Target Indication Key Features
AlloCAR-T (Allogene Therapeutics) BCMA, CD19 Multiple Myeloma, Leukemia Off-the-shelf CAR-T using CRISPR editing.
Poseida P-BCMA-101 BCMA Multiple Myeloma Stem cell memory T-cell platform for persistence.
SynNotch CAR-T Variable Solid Tumors Customizable T-cell activation via synNotch system.

 

CAR-T Therapy for Multiple Myeloma

 

Emerging Targets:

  • GPRC5D: A novel antigen with expression restricted to plasma cells.
  • FcRH5: Another emerging antigen for relapsed/refractory disease.

 

Combination Therapies:

CAR-T cells combined with immunomodulators, proteasome inhibitors, or monoclonal antibodies like daratumumab to enhance efficacy.

Next-generation CAR-T therapies are making strides in addressing the limitations of first-generation treatments. These advancements not only expand the range of cancers that can be treated but also improve patient outcomes through enhanced precision, safety, and accessibility.

Target Therapy

Disclaimer:- The content available on this website including text, graphics, logos is the property of TMM Hospital and is for informational and educational purposes only. All or any part of the material of this website shall not be copied, reproduced, distributed, modified, displayed, tampered, or used otherwise for any purpose without the prior written consent from the company.

TMM Hospital