New cancer treatment methods: Breakthrough

New cancer treatment methods: Breakthrough

I. Immunotherapy: Revolution in oncology

Immunotherapy, stimulating its own immune system of the body to combat cancer, has become one of the most significant achievements in oncology over the past decades. Unlike traditional methods, such as chemotherapy and radiation therapy that directly attack cancer cells, immunotherapy uses the power of the immune system to recognize and destroy malignant formations.

  • 1.1. Inhibitors of immune control points (Checkpoint Inhibitors)

    Inhibitors of immune control points, such as anti-CTLA-4, anti-PD-1 and anti-PD-L1, block proteins that prevent the activation of T cells. Under normal conditions, these control points prevent excessive immune reaction, but cancer cells often use them to avoid detection and destruction.

    • 1.1.1. The mechanism of action: CTLA-4, PD-1 and PD-L1 act as “brakes” on immune cells. CTLA-4 mainly acts in the early stages of T-cell activation in the lymph nodes, while PD-1/PD-L1 acts in peripheral tissues, in the micro-infection of the tumor. The blocking of these control points “removes the brakes” from the immune system, allowing T-cells to more effectively attack cancer cells.

    • 1.1.2. Clinical application: Inhibitors of immune control points showed impressive results in the treatment of various types of cancer, including melanoma, lung cancer, kidney cancer, Hodgkin lymphoma and bladder cancer. They can lead to prolonged remissions in patients who did not respond to other treatment methods.

    • 1.1.3. Side effects: Immunotherapy can cause side effects associated with hyperactivation of the immune system, known as immuno -mediated side effects (IRAES). These side effects may include inflammation of various organs, such as skin (dermatitis), intestines (colitis), liver (hepatitis), lungs (pneumonitis) and endocrine glands (pituitary, thyroiditis). Most IRAES can be effectively treated with corticosteroids and other immunosuppressive drugs.

  • 1.2. Cellular therapy (Cellular Therapy)

    Cell therapy includes the use of cells of the patient’s immune system or a cancer. There are several types of cell therapy, including CAR-T cell therapy, til therapy and therapy with NK cells.

    • 1.2.1. CAR-T CELLECTION ANTIGEN Receptor T-CELL Therapy): CAR-T cell therapy involves the genetic modification of the patient T-cells for the expression of a chimered antigenic receptor (CAR), which recognizes a specific antigen on the surface of cancer cells. After modification of CAR-T, the cells multiply in the laboratory and introduced back to the patient. Car-T cells then find and destroy cancer cells carrying the target antigen.

      • 1.2.1.1. The mechanism of action: CAR consists of an extracellular domain that recognizes a specific tumor antigen, a transmembrane domain and an intracellular domain that activates the T cells. When connecting Car with the antigen of the tumor, the T-cell is activated, which leads to its cytotoxic activity and the destruction of the cancer cell.

      • 1.2.1.2. Clinical application: Car-T cell therapy has shown remarkable results in the treatment of B-cell lymphomas and acute lymphoblastic leukemia (OLL) in children and adults. Several Car-T cell drugs are approved for the treatment of these diseases.

      • 1.2.1.3. Side effects: CAR-T cell therapy can cause serious side effects, including cytokine release syndrome (CRS) and neurotoxicity (icans). CRS is caused by the massive release of cytokines with activated Car-T cells and can manifest by fever, hypotension, hypoxia and organ dysfunction. ICANS can manifest itself in confusion, convulsions and a coma. These side effects require thorough monitoring and treatment.

    • 1.2.2. TIL Терапия (Tumor-Infiltrating Lymphocyte Therapy): Til Therapy involves the extraction of T cells that infiltrate the tumor from the patient’s tumor tissue. These T cells are reproduced in the laboratory and introduced back to the patient, often in combination with interleukin-2 (IL-2) to further stimulate their activity.

      • 1.2.2.1. The mechanism of action: Til is a population of T cells that have already recognized the antigens of the tumor and penetrated into tumor fabric. After propagation and introducing back to the patient, Til can more effectively attack the tumor.

      • 1.2.2.2. Clinical application: Til Therapy showed promising results in the treatment of melanoma, cervical cancer and other types of cancer.

      • 1.2.2.3. Side effects: Side effects of TIL therapy include fever, chills, hypotension and other side effects associated with the introduction of IL-2.

    • 1.2.3. Therapy NK NK cells are part of the congenital immune system and have the ability to destroy cancer cells without preliminary sensitization. Therapy with NK cells involves the use of NK cells of the patient or donor to combat cancer.

      • 1.2.3.1. The mechanism of action: NK cells recognize cancer cells by the absence of MHC class I molecules on their surface or by the expression of ligands that activate NK cells. After recognizing the cancerous cell, the NK cell releases cytotoxic granules that kill the cancer cage.

      • 1.2.3.2. Clinical application: Therapy with NK cells is studied in the treatment of various types of cancer, including leukemia, lymphoma and solid tumors.

      • 1.2.3.3. Side effects: Therapy with NK cells is usually well tolerated, but can cause side effects, such as fever, chills and cytokine storm.

  • 1.3. Oncolytic viruses (Oncolytic Viruses)

    Oncolytic viruses are viruses that selectively infect and destroy cancer cells without damaging healthy cells. They can be used both directly and for the delivery of therapeutic genes to cancer cells.

    • 1.3.1. The mechanism of action: Oncolytic viruses penetrate into cancer cells and multiply into them, causing cell lysis (destruction). During the lysis of cancer cells, viral particles that infect other cancer cells are released. In addition, oncolytic viruses can stimulate the immune system to attack cancer cells.

    • 1.3.2. Clinical application: T-VEC (Tallimogen Lagerparevpek) is an oncolytic virus approved for the treatment of melanoma. Oncolytic viruses are studied in the treatment of other types of cancer, including glioblasty, pancreatic cancer and ovarian cancer.

    • 1.3.3. Side effects: Side effects of oncolytic viruses are usually light and include flu -like symptoms, such as fever, chills and fatigue.

  • 1.4. Cancer Vaccines vaccines

    Cancer vaccines are designed to stimulate the immune system to recognize and attack cancer cells. There are two main types of cancer vaccines: preventive vaccines and therapeutic vaccines.

    • 1.4.1. Preventive vaccines: Preventive vaccines are designed to prevent the development of cancer caused by viruses. For example, a vaccine against the human papilloma virus (HPV) prevents cervical cancer, anus cancer and other types of cancer associated with HPV. Hepatitis B vaccine prevents liver cancer caused by hepatitis V. virus

    • 1.4.2. Therapeutic vaccines: Therapeutic vaccines are designed to treat existing cancer. They stimulate the immune system to the attack of cancer cells carrying specific antigens.

      • 1.4.2.1. The mechanism of action: Therapeutic vaccines contain tumor antigens that stimulate the immune system to the production of T cells and antibodies specific to cancer cells.

      • 1.4.2.2. Clinical application: Sipulerac-T is a therapeutic vaccine approved for the treatment of metastatic hormone-resistant cancer of the prostate gland. Therapeutic vaccines are studied in the treatment of other types of cancer, including melanoma, lung cancer and breast cancer.

      • 1.4.2.3. Side effects: Side effects of cancer vaccines are usually lung and include pain in the injection site, fever and fatigue.

II. Targeted therapy: accurate cancer blow

Targeted therapy is a type of cancer treatment that uses drugs aimed at specific molecules (for example, proteins) involved in the growth, progression and spread of cancer. Unlike chemotherapy, which attacks all rapidly dividing cells, targeted therapy is more selective and can have less side effects.

  • 2.1. Tyrosine Kinase Inhibitors – Tkis)

    Tyrosinkinase is enzymes that play a key role in transmitting signals inside cells, controlling the growth, differentiation and survival of cells. Tyrosinkinase inhibitors (TKIS) block the activity of these enzymes, thereby suppressing the growth and spread of cancer cells.

    • 2.1.1. The mechanism of action: TKIS bind to the active center of tyrosinkinase, preventing their phosphorylation and activation of lower signal tracts. This leads to inhibiting the growth and survival of cancer cells.

    • 2.1.2. Clinical application: TKIS are used to treat various types of cancer, including chronic myelolecosis (KHML), non -cancer lung cancer (NMRL), kidney cancer and gastrointestinal stromal tumors (GISO). Examples of TKIS include imatinib (glyc -beele), Gephitinib (Isses), Erlotinib (Tartseva) and Sunitinib (pimp).

    • 2.1.3. Side effects: The side effects of TKIS depend on a particular drug and can include leather rash, diarrhea, fatigue, nausea, vomiting and high blood pressure.

  • 2.2. Inhibitor mTOR (mTOR Inhibitors)

    MTOR (Michenen Rapamycin in mammals) is a protein that plays a key role in the regulation of growth, proliferation and cell metabolism. MTOR inhibitors block MTOR activity, thereby suppressing the growth and distribution of cancer cells.

    • 2.2.1. The mechanism of action: MTOR inhibitors bind to MTOR and inhibit its activity, blocking the lower signaling paths involved in the growth and proliferation of cells.

    • 2.2.2. Clinical application: MTOR inhibitors are used to treat kidney cancer, breast cancer and neuroendocrine tumors. Examples of MTOR inhibitors include Everolimus (Athenitor) and Temsirolimus (Torisel).

    • 2.2.3. Side effects: Side effects of MTOR inhibitors may include stomatitis, fatigue, nausea, vomiting, hyperglycemia and hyperlipidemia.

  • 2.3. CDK inhibitors (Cyclin-Dependent Kinase Inhibitors)

    CDK (cycle-dependent kinase) is enzymes that play a key role in the regulation of the cell cycle. CDK inhibitors block the activity of CDK, thereby stopping the cell cycle and suppressing the growth of cancer cells.

    • 2.3.1. The mechanism of action: CDK inhibitors bind to CDK and inhibit their activity, preventing the phosphorylation of proteins necessary for the progression of the cell cycle.

    • 2.3.2. Clinical application: CDK4/6 inhibitors are used to treat hormone-positive, Her2-negent breast cancer. Examples of CDK4/6 inhibitors include Palbocyclib (Ibrans), Ribocyclib (Kisgali) and Abemaciplib (Verrasenio).

    • 2.3.3. Side effects: Side effects of CDK4/6 inhibitors may include neutropenia, fatigue, nausea and diarrhea.

  • 2.4. Parp Inhibitors (Parp Inhibitors)

    PARP (Paul (Adf Ribose)-Polymerase) is an enzyme that plays a key role in DNA restoration. PARP inhibitors block PARP activity, thereby preventing DNA restoration in cancer cells, especially in cells with defects in BRCA1 or BRCA2 genes.

    • 2.4.1. The mechanism of action: PARP inhibitors are associated with PARP and inhibit its activity, preventing the restoration of damaged DNA. In cells with defects in the BRCA1 or BRCA2 genes, which also participate in DNA restoration, this leads to the accumulation of DNA damage and cell death.

    • 2.4.2. Clinical application: PARP inhibitors are used to treat ovarian cancer, breast cancer, prostate cancer and pancreatic cancer in patients with mutations in BRCA1 or BRCA2 genes. Examples of PARP inhibitors include Olaparib (Lindparza), Rubaparib (Rubraka) and Talazoparib (Talzenna).

    • 2.4.3. Side effects: Side effects of PARP inhibitors may include nausea, fatigue, vomiting, anemia and thrombocytopenia.

  • 2.5. Monoclonal antibodies – Mabs)

    Monoclonal antibodies are antibodies that specifically associate with certain antigens on the surface of cancer cells. They can be used for various purposes, including blocking signal tracks, stimulating the immune system and delivering toxic substances to cancer cells.

    • 2.5.1. The mechanism of action: Monoclonal antibodies can act in several ways. They can block receptors on the surface of cancer cells, preventing ligand binding and activating signal routes. They can stimulate the immune system to attack cancer cells by connecting antigens on the surface of cancer cells and activating a complement or attracting immune cells. They can deliver toxic substances, such as chemotherapeutic drugs or radioactive isotopes, to cancer cells.

    • 2.5.2. Clinical application: Monoclonal antibodies are used to treat various types of cancer, including breast cancer, lung cancer, colon cancer and lymphoma. Examples of monoclonal antibodies include trastuzumab (heceptin), Bevacizumab (Avastin), Rituximab (Mabter) and Cetuximab (Erbitux).

    • 2.5.3. Side effects: Side effects of monoclonal antibodies depend on a particular drug and may include infusion reactions, skin rash, diarrhea and fatigue.

III. Genomic profiling: a personalized approach to treatment

Genomal profiling, also known as molecular profiling, includes an analysis of DNA, RNA and proteins of the patient’s cancer cells to detect genetic mutations and other molecular changes, which can be used to select the most effective treatment. This personalized approach to treatment allows doctors to prescribe medications that are more likely to be effective and will be less likely to cause side effects.

  • 3.1. New generation sequencing (NEXT -GENERATION SEQUENCING – NGS)

    A new generation sequencing (NGS) is a technology that allows you to quickly and inexpensively secure large areas of DNA or RNA. NGS is used to identify genetic mutations, amplification and delections in cancer cells.

    • 3.1.1. The mechanism of action: NGS includes fragmentation of DNA or RNA, amplification of fragments and sequencing of fragments. The resulting sequences are then compared with a reference genome to detect genetic changes.

    • 3.1.2. Clinical application: NGS is used to detect drive -up mutations in cancer cells, which can be targets for targeted therapy. For example, NGS can be used to identify mutations in EGFR, Alk or ROS1 genes in mercilecock lung cancer, which can be targets for TKIS. NGS can also be used to identify mutations in the BRCA1 or BRCA2 genes, which can make cancer more susceptible to PARP inhibitors.

    • 3.1.3. Advantages: NGS allows you to simultaneously analyze many genes, which makes it more effective and economical than traditional sequencing methods.

  • 3.2. Immunohistochemistry (Immunohistochemastry – IHC)

    Immunohistochemistry (IHC) is a method that uses antibodies to detect certain proteins in tissue. IHC is used to determine the expression of proteins that can be targets for targeted therapy or which may indicate sensitivity or resistance to certain types of treatment.

    • 3.2.1. The mechanism of action: IHC includes incubation of fabric with antibodies specific to a certain protein. Then the antibody is associated with protein, and binding is detected using an enzymatic or fluorescent reaction. The staining intensity indicates the level of protein expression.

    • 3.2.2. Clinical application: IHC is used to determine the expression of estrogen and progesterone receptors (ER/PR) and HER2 in breast cancer. ER/PR-positive breast cancer can be treated with hormonal therapy, and Her2-positive breast cancer can be treated with trastuzumab. IHC is also used to determine PD-L1 expression in lung cancer, which may indicate the probability of response to immune control points inhibitors.

  • 3.3. Liquid biopsy (Liquid Biopsy)

    Liquid biopsy is a method that involves the analysis of blood samples or other body fluids to detect cancer cells, DNA or other molecules released from the tumor. Liquid biopsy can be used to monitor the response to treatment, detecting cancer relapse and detect targets for targeted therapy.

    • 3.3.1. The mechanism of action: Liquid biopsy may include the release of circulating tumor cells (CTC), circulating tumor DNA (CTDNA) or exosia from a blood sample. CTC is cancer cells that separated from the tumor and circulate in the blood. CTDNA is a DNA released from cancer cells into the blood. Exosomas are small bubbles that are released from cells and contain proteins, RNA and DNA.

    • 3.3.2. Clinical application: Liquid biopsy can be used to detect genetic mutations in CTDNA, which can be targets for targeted therapy. Liquid biopsy can also be used to monitor the CTDNA level during treatment to evaluate the response to treatment and identify cancer relapse.

IV. Radiation therapy: Improving the methods

Radiation therapy uses high -energy rays to destroy cancer cells. Modern methods of radiation therapy allow you to more accurately direct radiation to the tumor, minimizing damage to the surrounding healthy tissues.

  • 4.1. Stereotactic radiation therapy.

    Stereotactic radiation therapy (SRT) is a type of radiation therapy that uses high -precision guidance methods to deliver high doses of radiation to small tumors. SRT can be used to treat tumors in the brain, lungs, liver and other organs.

    • 4.1.1. The mechanism of action: SRT uses a stereotactic frame or other guidance methods to accurately determine the position of the tumor. Then the radiation is delivered to the tumor from different directions, which allows you to deliver a high dose of radiation to the tumor, minimizing damage to the surrounding healthy tissues.

    • 4.1.2. Types SRT: There are two main types of SRT: stereotactic radiosurgery (SRS) and stereotactic radiation therapy (SBRT). SRS is usually used to treat tumors in the brain, while SBRT is usually used to treat tumors in other organs.

    • 4.1.3. Advantages: SRT allows you to deliver high doses of radiation to the tumor, which can increase the effectiveness of treatment. SRT also minimizes damage to the surrounding healthy tissues, which can reduce side effects of treatment.

  • 4.2. Proton therapy (Proton Therapy)

    Proton therapy is a type of radiation therapy that uses protons (positively charged particles) instead of photons (x -rays). Protons have unique physical properties that allow them to deliver most of their energy to the tumor and minimally damage the surrounding healthy tissues.

    • 4.2.1. The mechanism of action: Protons have a certain depth of penetration, which depends on their energy. After reaching this depth, protons release most of their energy, which is known as the Bragg Peak. This allows you to deliver a high dose of radiation to the tumor, minimizing tissue damage located before and after the tumor.

    • 4.2.2. Advantages: Proton therapy can reduce the side effects of treatment and increase the effectiveness of treatment, especially in the treatment of tumors located near critical organs.

  • 4.3. Brachytherapy

    Brachitherapy is a type of radiation therapy, which involves the placement of a radioactive source directly inside or next to the tumor. Brachitherapy allows you to deliver a high dose of radiation to the tumor, minimizing damage to the surrounding healthy tissues.

    • 4.3.1. The mechanism of action: A radioactive source is placed inside or next to the tumor using applicators or needles. The radiation emitted by the source destroys cancer cells.

    • 4.3.2. Clinical application: Brachitherapy is used to treat various types of cancer, including prostate cancer, cervical cancer, breast cancer and skin cancer.

V. Surgery: Miniyinvasive methods and robotics

Surgery still plays an important role in the treatment of many types of cancer. Modern surgical methods, such as mini -vinvasive surgery and robotic surgery, allow you to remove tumors with less damage to surrounding tissues, which leads to faster restoration and reduction of complications.

  • 5.1. Laparoscopic surgery.

    Laparoscopic surgery, also known as Miniynvasive Surgery, involves the operation through small incisions using a special tool called the laparoscope. Laparoscope is a thin tube with a camera at the end, which allows the surgeon to see the internal organs on the monitor screen.

    • 5.1.1. The mechanism of action: The surgeon makes several small cuts in the skin and introduces a laparoscope and other surgical instruments through these cuts. The surgeon uses tools for removing the tumor and other affected fabrics.

    • 5.1.2. Advantages: Laparoscopic surgery has several advantages compared to open surgery, including less pain, a shorter stay in the hospital, faster recovery and smaller scars.

  • 5.2. Robotic Surgery.

    Robotized surgery is a type of laparoscopic surgery that uses a surgical robot to perform surgery. The surgeon controls the robot using a console, which provides a three -dimensional image of the operating field and allows the surgeon to perform complex movements with greater accuracy and control.

    • 5.2.1. The mechanism of action: The surgeon sits behind the console and controls the hands of the robot that hold surgical instruments. The robot repeats the movement of the surgeon, but with greater accuracy and stability.

    • 5.2.2. Advantages: Robotized surgery has several advantages compared to traditional laparoscopic surgery, including more accurate movements, an improved review of the operating field and a more comfortable position for the surgeon.

  • 5.3. Intraoperative radiation therapy.

    Intraoperative radiation therapy (IRT) is a type of radiation therapy, which is carried out directly during the operation after removal of the tumor. IRT allows you to deliver a high dose of radiation to the remains of a tumor or areas that are at risk of relapse, minimizing damage to surrounding healthy tissues.

    • 5.3.1. The mechanism of action: After removing the tumor, the surgeon placed an applique or electronic radiation source in the operating field. Then radiation is delivered to the fabrics surrounding the operating field.

    • 5.3.2. Clinical application: IRT is used to treat various types of cancer, including breast cancer, stomach cancer and pancreatic cancer.

VI. Future of cancer treatment: new horizons

Studies in the field of cancer treatment continue to develop rapidly, opening new horizons and offering hope for more effective and less toxic treatment methods.

  • 6.1. Adaptive therapy (Adaptive Therapy)

Adaptive therapy is a treatment strategy that involves the adaptation of the dose and schedule of drug administration depending on the tumor response to treatment. The purpose of adaptive therapy is to control the growth of the tumor, and not its complete destruction, which can reduce the development of resistance to drugs.

  • 6.2. Artificial intelligence (artificial intelligence – ai) in oncology

Artificial intelligence (AI) plays an increasingly important role in oncology. AI can be used to analyze medical images, identify genetic mutations, predict a response to the treatment and development of new drugs.

  • 6.3. Nanotechnology in the treatment of cancer

Nanotechnologies offer new opportunities for the delivery of drugs directly to cancer cells, increase the effectiveness of treatment and reduce side effects. Nanoparticles can be developed for specific binding with cancer cells and the release of drugs only in these cells.

  • 6.4. Personalized cancer vaccines.

Personalized cancer vaccines are developed on the basis of the genetic profile of the patient’s tumor. These vaccines stimulate the immune system to the attack of cancer cells that carry specific antigens present only in the patient’s tumors.

These new methods of cancer treatment and ongoing research open new horizons in the fight against this disease and offer hope for more effective and high -quality treatment in the future.

Leave a Reply

Your email address will not be published. Required fields are marked *