Breakthrough in the treatment of cancer: new hopes for patients
Section 1: Revolution in immunotherapy
1.1. The basics of cancer immunotherapy
Immunotherapy, in contrast to traditional treatment methods, such as chemotherapy and irradiation, which directly attack cancer cells, is aimed at mobilizing the patient’s immune system to combat cancer. It uses its own protective mechanisms of the body to recognize and destroy tumor cells. This opens up new horizons in the treatment of cancer, since the immune system has the ability to adapt and remember cancer cells, potentially providing long -term control over the disease and preventing it from relapse.
Immunotherapy operates in several ways:
- Blocking control points of immunity: Cancer cells often use mechanisms that suppress the immune system to avoid detection and destruction. Immunotherapy, in particular control points inhibitors, blocks these mechanisms, allowing immune cells such as T-lymphocytes, to attack cancer.
- Adaptive cell therapy: This approach includes the extraction of immune cells from the patient’s body, a genetic modification to increase their ability to recognize and attack cancer cells, the reproduction of these cells in the laboratory and return to the patient.
- Cancer vaccines: They stimulate the immune system to recognize and attack specific cancer cells. Unlike preventive vaccines that prevent infection, therapeutic cancer vaccines are aimed at treating existing cancer.
- Immunomodulator: These are substances that stimulate or restore the immune system, increasing its ability to fight cancer.
1.2. Control points inhibitors: paradigm change
Control points (Checkpoint Inhibitors – CPI) are a class of immunotherapeutic drugs that revolutionized many types of cancer. These drugs act, blocking proteins-control points that regulate the activity of T-lymphocytes. By blocking these control points, CPI remove the “brakes” from the immune system, allowing T-lymphocytes to more effectively attack cancer cells.
The most famous CPI includes:
- CTLA-4 inhibitors (Cytotoxic T-Lymphocyte-SSOSOCIATED PROTEIN 4): CTLA-4 is a control point that reduces the activity of T-lymphocytes in the early stages of activation. Blocking CTLA-4 promotes a stronger activation of T-lymphocytes and enhances their ability to destroy cancer cells.
- PD-1 inhibitors (Programmed Cell Death Protein 1): PD-1 is a control point that reduces the activity of T-lymphocytes in the tumor tissue. Blocking PD-1 allows T-lymphocytes to more efficiently attack cancer cells located in the micro-infection of the tumor.
- PD-L1 inhibitors (Programmed Death-Ligand 1): PD-L1 is a protein expressed on cancer cells, which binds to PD-1 on T-lymphocytes, suppressing their activity. Blocking PD-L1 prevents this interaction and allows T-lymphocytes to attack cancer cells.
Examples of CPI approved for the treatment of various types of cancer:
- Yeilimumab (iPilimumab): CTLA-4 inhibitor is used to treat melanoma, kidney cancer and lung cancer.
- Pembrolizumab (Pembrolizumab): PD-1 inhibitor is used to treat melanoma, lung cancer, Hodgkin lymphoma and other types of cancer.
- Nivolumab (nivolumab): PD-1 inhibitor is used to treat melanoma, lung cancer, kidney cancer, Hodgkin lymphoma and other types of cancer.
- Atezolizumab (atzolizumab): PD-L1 inhibitor is used to treat lung cancer, bladder cancer and other types of cancer.
CPI showed significant efficiency in the treatment of various types of cancer, which were previously considered incurable. However, they can also cause side effects associated with hyperactivation of the immune system, such as autoimmune diseases.
1.3. Adaptive cell therapy: personalized approach
Adaptive cell therapy (ACT) is a personalized approach to immunotherapy, which uses the patient’s own immune cells to combat cancer. ACT includes the extraction of T-lymphocytes from the patient’s body, their genetic modification to increase their ability to recognize and attack cancer cells, the reproduction of these cells in the laboratory and return to the patient.
The most promising ACT forms include:
- CAR-T CELLECTION ANTIGEN Receptor T-CELL Therapy): CAR-T cell therapy includes the genetic modification of the patient’s T-lymphocytes using a chimeric antigenic receptor (CAR), which allows them to recognize and attack specific proteins present on the surface of cancer cells. Car-T cells are introduced back to the patient and, detecting cancer cells, activate and destroy them. CAR-T cell therapy has shown high efficiency in the treatment of certain types of leukemia and lymphoma.
- TIL терапия (Tumor-infiltrating lymphocyte therapy): Til Therapy includes the extraction of T-lymphocytes, which have already entered the tumor (Til), from the patient’s tumor. These til are propagated in the laboratory and then introduced back to the patient. Til Therapy uses the natural ability of the immune system to recognize and attack cancer cells, but enhances this ability by propagating and activating TIL outside the patient’s body. Til Therapy has shown the effectiveness in the treatment of melanoma and other solid tumors.
CAR-T cell therapy and til therapy are complex and expensive treatment methods, but they demonstrate significant potential in cancer treatment.
1.4. Cancer-specific vaccines: training of the immune system
Cancer vaccines are designed to stimulate the immune system to recognize and attack specific cancer cells. Unlike preventive vaccines that prevent infection, therapeutic cancer vaccines are aimed at treating existing cancer.
Cancer-specific vaccines can be developed for aiming at:
- Cancer antigens: These are proteins that are specifically expressed in cancer cells, but not normal cells. Vaccines aimed at cancer antigens stimulate the immune system to recognize and attack cells expressing these antigens.
- Neoantigens: These are new antigens that occur as a result of mutations in cancer cells. Neoantigens are unique to each tumor and can be used to develop personalized vaccines.
Cancer-specific vaccines can be made of:
- Cancer cells: Weakened or killed cancer cells can be used to stimulate the immune system.
- Cancer antigens or neoantigen: Synthetic peptides, proteins or DNA, encoding cancer antigens or neoantigens can be used to stimulate the immune system.
- Dendritic cells: Dandrit cells are immune cells that represent antigens of T-lymphocytes. Dandrit cells can be loaded with cancer antigens or neoantigens in the laboratory and then introduced back to the patient to stimulate the immune response.
Cancer-specific vaccines show promising results in clinical trials, especially in combination with other types of immunotherapy.
1.5. Combinations of immunotherapy: synergistic effect
Combinations of immunotherapy make it possible to achieve a synergistic effect, enhancing the immune response against cancer. The combination of various immunotherapeutic approaches can overcome the mechanisms of cancer resistance and improve clinical outcomes.
Examples of combinations of immunotherapy:
- Ingibitors of Ingibitors CTLA-4 and PD-1: Simultaneous blocking CTLA-4 and PD-1 can enhance the activation of T-lymphocytes and increase their ability to attack cancer cells.
- Combination of immunotherapy and chemotherapy: Chemotherapy can destroy cancer cells and release cancer antigens that can be used by the immune system to develop an immune response. Immunotherapy can strengthen this immune response and help destroy the remaining cancer cells.
- Combination of immunotherapy and radiation therapy: Radiation therapy can damage cancer cells and release cancer antigens that can be used by the immune system to develop an immune response. Immunotherapy can strengthen this immune response and help destroy the remaining cancer cells.
- Combination of immunotherapy and targeted therapy: Targeted therapy can be aimed at specific molecules, which are important for the growth and survival of cancer cells. Immunotherapy can enhance the effect of targeted therapy and help destroy cancer cells that become resistant to targeted therapy.
Combinations of immunotherapy are a promising approach to cancer treatment, but they can also cause more serious side effects.
Section 2: Targeted therapy: Awear Back to Cancer
2.1. The principles of targeted therapy
Targeted therapy is a class of anti -cancer drugs that are aimed at specific molecules that are important for growth, progression and survival of cancer cells. Unlike chemotherapy, which affects all rapidly dividing cells (including healthy), targeted therapy is aimed at specific molecular goals present in cancer cells. This leads to a more purposeful and selective destruction of cancer cells, which can reduce side effects.
Targeted therapy acts in several ways:
- Signaling track locks: Cancer cells often use aberrant signaling paths to stimulate growth and division. Targeted drugs can block these signaling paths, depriving cancer cells of the necessary signals for growth and survival.
- Angiogenesis inhibiting: Cancer tumors need new blood vessels to provide oxygen and nutrients. Targeted drugs can inhibit angiogenesis, depriving the tumor of the resources necessary for growth.
- Induction apoptosis: Targeted drugs can induce apoptosis (programmed cell death) in cancer cells, causing their self -destruction.
- Delivery of toxic substances directly to cancer cells: Some targeted drugs are associated with toxic substances, such as chemotherapeutic drugs or radioisotopes. The targeting part of the drug directs toxic substance directly to cancer cells, minimizing damage to healthy cells.
2.2. Tyrosinkinase inhibitors: growth blocking
Tyrosinkinase inhibitors (TKI) are a class of targeted drugs that block the activity of tyrosinkinase, enzymes that play a key role in transmitting cell growth and cell division. Many cancer cells express aberrant tyrosinkinase, which stimulate uncontrolled growth and proliferation. TKI block these aberrant tyrosinkinase, depriving cancer cells of the necessary signals for growth and survival.
Examples of TKI used to treat various types of cancer:
- Imatinib (imatinib): BCR-BL tyrosinkinase inhibitor is used to treat chronic myelolecosis (KML).
- Gefitinib (Gefitinib) and Erlotinib (Erlotinib): EGFR tyrosynase inhibitors are used to treat lung cancer, non -alcohol -type (NSCLC).
- Sorafenib (Sunitinib): Sorafenib: Multiple tyrosinkinase inhibitors are used to treat kidney cancer and liver cancer.
- Crizotinib): ALK tyrosinkinase inhibitor is used to treat lung cancer, non -cell -type type (NSCLC) with Alk mutation.
TKI showed significant efficiency in the treatment of various types of cancer, especially those characterized by specific genetic mutations leading to aberrant activation of tyrosyankinase.
2.3. MTOR inhibitors: Cancer metabolism control
MTOR inhibitors (Michenen Rapamycin in mammals) are a class of targeted drugs that block the activity of MTOR, proteinquinase, which plays a key role in the regulation of cell growth, proliferation, metabolism and angiogenesis. MTOR is the central node of several signaling pathways that control cell growth and survival, and its aberrant activation is often observed in cancer cells.
Examples of MTOR inhibitors used to treat various types of cancer:
- Everolimus (Everolimus): It is used to treat kidney cancer, breast cancer and pancreatic tumors.
- Temsirolimus (Temsirolimus): Used to treat kidney cancer.
MTOR inhibitors showed the effectiveness in the treatment of various types of cancer, especially in combination with other types of targeted therapy or hormonal therapy.
2.4. Monoclonal antibodies: accurate delivery
Monoclonal antibodies (MAB) are a class of targeted drugs, which are antibodies created in laboratories for recognition and binding with specific proteins present on the surface of cancer cells. After binding with cancer cells, MAB can act in several ways:
- Signaling track locks: MAB can block the signaling paths of growth and proliferation, blocking the binding of ligans with receptors on the surface of cancer cells.
- Induction apoptosis: Mab can induce apoptosis in cancer cells, associated with receptors on their surface.
- Activation of the immune system: Mab can attract immune cells to cancer cells, which leads to their destruction.
- Delivery of toxic substances: MAB can be associated with toxic substances such as chemotherapeutic drugs or radioisotopes, and deliver them directly to cancer cells.
Examples of MAB used to treat various types of cancer:
- Trastuzumab (Trastuzumab): aimed at the HER2 protein, used to treat breast cancer with high expression HER2.
- Rituximab (Rituximab): aimed at CD20 protein, used to treat lymphoma.
- Bevacizumab (Bevacizumab): Aims at the growth factor of the vascular endothelium (VEGF), used to treat lung cancer, colon cancer and kidney cancer.
- Cetuximab (Cetuximab): Aims at the receptor of the epidermal growth factor (EGFR), used to treat cancer of the colon and cancer of the head and neck.
Mab has shown significant efficiency in the treatment of various types of cancer, especially in combination with other types of therapy.
2.5. PARP Ingitors: Operation of cancer weaknesses
PARP Ingators (Polie inhibitors (ADF Ribose)-polymerase) are a class of targeted drugs that block the activity of PARP, an enzyme that plays a key role in DNA reparation. Cancer cells with defects in other DNA reparations, such as BRCA1 and BRCA2, are especially sensitive to PARP Ingibitors. Blocking PARP in these cells leads to the accumulation of DNA damage and cell death.
Examples of PARP Ingitors used to treat various types of cancer:
- Showing (gearly): Used to treat ovarian cancer, breast cancer and prostate cancer with BRCA1/2 mutations.
- Rucaparib (Rucaparib): Used to treat ovarian cancer with BRCA1/2 mutations.
- Talazoparib (Talazoparib): Used to treat breast cancer with BRCA1/2 mutations.
PARP Ingators showed the effectiveness in the treatment of cancer tumors with defects in DNA reparation, especially BRCA1/2-associated crayfish.
Section 3: Genomic sequencing and personalized medicine
3.1. The role of genomic sequencing in the treatment of cancer
Genomic sequencing, the process of determining the complete sequence of DNA of the patient’s cancer cells, has become a powerful tool in personalized medicine in oncology. It allows you to identify genetic mutations that lead to the growth and progression of cancer, and also determines which targeted therapy or immunotherapy is most likely to be effective for a particular patient.
Genomic sequencing can be performed on samples of tumor tissue or on liquid biopsy (blood sample containing cancer or DNA). Information obtained as a result of genomic sequencing is used for:
- Cancer diagnostics: Genomic sequencing can help in cancer diagnosis, especially in cases where traditional diagnostic methods do not give unambiguous results.
- Prediction of the outcome of cancer: Genomic sequencing can help predict how cancer will progress and how the patient will react to treatment.
- The choice of the most effective treatment: Genomic sequencing can help doctors choose the most effective treatment for a particular patient, based on the genetic profile of cancer.
- Monitoring of the response to treatment: Genomic sequencing can be used to monitor the response of cancer for treatment and to identify the development of resistance.
3.2. Identification of driver mutations
Driver mutations are genetic changes that directly contribute to the growth and progression of cancer. The identification of driver mutations is crucial for the development of targeted therapy, which is aimed at these specific mutations.
Examples of driver mutations, which are often detected during genomic sequencing:
- EGFR (receptor of the epidermal growth factor): Mutations in the EGFR gene are often found with lung cancer, non -alcohol type (NSCLC) and are a target for TKI, such as Gephitinib and Erlotinib.
- Alk (kinaza lymphomas of anaplastic): Perestroika in the ALK gene is often found with lung cancer, non -cell -type (NSCLC) and are a target for TKI, such as crisotinib.
- Braf (b-raf ProtooCogen): Mutations in the BRAF gene are often found with melanoma and are a target for BRAF inhibitors, such as Vemurafenib and Dubrafenib.
- Pik3CA (phosphatidydilinositol-4.5-bisfosphate 3-kinase, catalytic subunit alpha): Mutations in Pik3CA gene are often found in breast cancer and are a target for PI3K inhibitors.
- KRAS (Kirsten rat sarcoma viral oncogene homolog): Mutations in the KRAS gene are often found with cancer of the colon and lung cancer, but for a long time were considered inaccessible to targeted therapy. However, in recent years, new drugs have been developed that can aim at KRAS mutations.
3.3. Biomarkers for immunotherapy
Genomic sequencing can also be used to identify biomarkers, which predict the likelihood of response to immunotherapy.
Examples of biomarkers for immunotherapy:
- PD-L1 (Ligand of Programed Death-1): PD-L1 expression on cancer cells may predict the probability of response to PD-1/PD-L1 inhibitors.
- TMB (mutation load of the tumor): TMB is the number of mutations in DNA of cancer cells. Higher TMB is more likely to respond to immunotherapy, since cancer cells with a large number of mutations are more likely expressed by non -antigens that can be recognized by the immune system.
- MSI (microsateline instability): MSI is a condition in which there is a large number of mutations in microsatellite areas in the DNA of cancer cells. Crayfish with high MSI is more likely to respond to immunotherapy.
3.4. Clinical trials based on genomic profiling
Genomal profiling has led to the development of clinical tests based on the genetic profile of cancer tumors of patients. In these clinical trials, patients receive treatment based on specific genetic mutations of their cancer tumors, and not on the type of cancer. This allows doctors to be more effectively aimed at cancer and improve clinical outcomes.
Examples of clinical trials based on genomic profiling:
- NCI-MATCH (National Cancer Institute Molecular Analysis for Therapy Choice): This is a large clinical test in which patients with various types of cancer receive treatment based on genetic mutations of their cancer tumors.
- TAPUR (Targeted Agent and Profiling Utilization Registry): This is a clinical test in which patients with progressive cancer receive treatment with Targeted drugs based on the genetic profile of their cancerous tumors.
3.5. Obstacles and prospects for personalized medicine
Personalized medicine in oncology has great potential to improve cancer treatment, but there are also obstacles to its wide implementation.
Obstacles for personalized medicine:
- High cost of genomic sequencing: Genomic sequencing is still expensive, which limits its accessibility for many patients.
- The difficulty of interpretation of genomic data: The interpretation of genomic data requires special knowledge and experience.
- Lack of targeted therapy for all driver mutations: Targeted therapies are not available for all driverships.
- The development of resistance to targeted therapy: Cancer cells can over time develop resistance to targeted therapy.
Prospects for personalized medicine:
- Reducing the cost of genomic sequencing: The cost of genomic sequencing is reduced, which makes it more affordable for patients.
- Development of new targeted therapy: New targeted therapies are being developed, which are aimed at more drivers.
- Development of strategies to overcome resistance to targeted therapy: Strategies are developed to overcome resistance to targeted therapy, such as combined therapy and the use of new generation inhibitors.
- The wider use of clinical trials based on genomic profiling: Clinical trials based on genomic profiling are becoming increasingly common, which allows doctors to more effectively aim at cancer and improve clinical outcomes.
Section 4: New horizons in cancer treatment technologies
4.1. CRISPR-CAS9: Genomic editing
CRISPR-CAS9 (Clustered Regularly Interspaced Short Palindromic Repeats-CRISPR-SSOSOCIETED Protein 9) is a revolutionary genomic editing technology that allows scientists with high accuracy to change DNA of living organisms. In the context of treatment CRISPR-CAS9 cancer can be used for:
- Inactivations oncogenov: Oncogenes are genes that contribute to the growth and progression of cancer. CRISPR-CAS9 can be used to inactivation of oncogenes, which can stop the growth of cancer.
- Restoration of tumor-soup genes: Tumor-soup genes are genes that prevent cancer growth. CRISPR-CAS9 can be used to restore tumor-soup genes, which can stop the growth of cancer.
- Creating CAR-T cells: CRISPR-CAS9 can be used to create CAR-T cells, which are more efficient on cancer cells.
- Development of new cancer diagnostics: CRISPR-CAS9 can be used to develop new cancer diagnostics, which are more accurate and sensitive.
CRISPR-CAS9 is a promising technology that has the potential to revolutionize cancer treatment, but it is still in the early stages of development. It is necessary to conduct additional studies to assess the safety and effectiveness of CRISPR-CAS9 in the treatment of cancer.
4.2. Nanotechnology in the treatment of cancer
Nanotechnologies are the use of materials and devices on the nanotherapy (one billionth meter). In the context of treatment of nanotechnology cancer, can be used for:
- Targeted delivery of drugs: Nanoparticles can be developed for targeted delivery of drugs directly to cancer cells, which can increase the effectiveness of treatment and reduce side effects.
- Visualization of cancer tumors: Nanoparticles can be used to visualize cancer tumors, which can help doctors diagnose cancer in the early stages and control the response to treatment.
- Hyperthermia: Nanoparticles can be used to heat cancer cells, which can lead to their destruction.
- Photodynamic therapy: Nanoparticles can be used to deliver photosensitive substances to cancer cells. Then light radiation is used to activate these substances, which leads to the destruction of cancer cells.
Nanotechnology is a promising field of research, which has the potential to significantly improve cancer treatment. It is necessary to conduct additional studies to assess the safety and effectiveness of nanotechnologies in the treatment of cancer.
4.3. Liquid biopsy: non -invasive monitoring
Liquid biopsy is a non -invasive cancer monitoring method, which includes a patient’s blood test to detect cancer cells or DNA released by cancer cells. Liquid biopsy has several advantages over traditional tumor biopsy:
- Non -invasiveness: Liquid biopsy is non -invasive, which means that it does not require surgical intervention.
- The possibility of multiple repetition: The liquid biopsy can be repeated many times, which allows doctors to control the response to treatment in real time.
- Obtaining information about the genetic heterogeneity of the tumor: Liquid biopsy can provide information about the genetic heterogeneity of the tumor, which can help doctors choose the most effective treatment.
- Identification of the minimum residual disease: Liquid biopsy can be used to detect a minimum residual disease (MRB), that is, a small amount of cancer cells that remain after treatment. The identification of MRB can help doctors decide on the need to conduct additional treatment.
Liquid biopsy is a promising tool for monitoring cancer, which can improve clinical outcomes. It is necessary to conduct additional research to assess its effectiveness in various clinical situations.
4.4. Artificial intelligence and machine learning in oncology
Artificial intelligence (AI) and machine learning (MO) are rapidly developing and have an increasing influence on oncology. AI and MO can be used for:
- Cancer diagnostics: AI and MO can be used to analyze medical images (for example, x -rays, KT and MRI) to identify cancer tumors.
- Forecasting the outcome of cancer: AI and MO can be used to predict the outcome of cancer based on clinical data and the patient’s genomic profile.
- Selection of the most effective treatment: AI and MO can be used to select the most effective treatment for a particular patient based on clinical data, genomic cancer profile and drug sensitivity data.
- Development of new drugs: AI and MO can be used to develop new drugs for cancer by identifying new medicinal purposes and predicting the effectiveness of new drugs.
AI and MO have great potential for converting oncology. It is necessary to conduct additional research to develop and implement these technologies into clinical practice.
4.5. Proton therapy: exact irradiation
Proton therapy is a type of radiation therapy that uses protons (positively charged particles) instead of x -rays to destroy cancer cells. Proton therapy has several advantages over traditional radiation therapy:
- More accurate irradiation: Protons can be directed with greater accuracy at the tumor, which allows you to reduce the radiation dose obtained by healthy tissues.
- Less side effects: Proton therapy causes less side effects than traditional radiation therapy.
- The possibility of treating crayfish, which are inaccessible to surgical intervention: Proton therapy can be used to treat crayfish, which are difficult to access for surgical intervention or are located near the sensitive organs.
Proton therapy is an effective method of treating cancer, but it is available only in a few centers around the world.
Section 5: Psychological support and quality of life
5.1. The importance of psychological support
The diagnosis of cancer has a huge impact not only on the physical, but also on the psychological state of the patient. Fear, anxiety, depression, a feeling of isolation and helplessness are only some of the emotions that cancer patients face and their loved ones. Psychological support plays a key role in adaptation to the disease, reducing stress, improving the quality of life and increasing the effectiveness of treatment.
Psychological support helps patients:
- Cope with emotional stress: Psychologists help patients understand their feelings, learn to cope with anxiety, fear and depression.
- Improve communication with doctors and loved ones: Psychologists teach patients effective communication techniques so that they can better understand the information provided by doctors, and express their needs and fears to their loved ones.
- Strengthen self -confidence and your abilities: Psychologists help patients realize their strengths, develop strategies for overcoming difficulties and increase self -confidence and their abilities.
- Take the disease and adapt to changes: Psychologists help patients take a disease, adapt to changes in their lives and find new goals and meanings.
- Improve the quality of life: Psychologists help patients improve the quality of life, despite the disease, by increasing their emotional well -being, social activity and physical activity.
5.2. Types of psychological assistance
There are various types of psychological care, accessible for patients with cancer and their loved ones:
- Individual psychotherapy: Individual psychotherapy allows patients to work one on one with a psychologist to solve their personal problems and develop strategies for overcoming difficulties.
- Group psychotherapy: Group psychotherapy allows patients to communicate with other people faced with similar problems, share their experience and support each other.
- Family psychotherapy: Family psychotherapy allows families to work together with a psychologist to solve the problems associated with cancer, and improve communication and understanding.
- Consultations on psychological health: Consultations on psychological health provide patients with information about cancer, its treatment and side effects, as well as Coping strategies and support resources.
- Relaxation techniques: Relaxation techniques, such as meditation, yoga and progressive muscle relaxation, can help patients reduce stress, anxiety and pain.
5.3. Family support and loved ones
Cancer is a disease that affects not only the patient, but also his family and loved ones. Family and relatives also need psychological support to cope with emotional stress, anxiety and a sense of helplessness.
Support for family and loved ones includes:
- Providing information: Providing information about cancer, its treatment and side effects, and