top of page
Archive

Insight of Electric Fields Therapy

These articles section encompasses more than just articles about ECCT. It aimed to educate and provide insights about general electric field therapy application from a wide array of source worldwide, including news, opinions, research findings, educational content, professional advice, and more. 

Section Title

This paper explores how electric fields can help treat cancer. It shows that electric fields disrupt cancer cell division, making the cells die and stopping them from spreading. They also enhance the immune system's ability to fight cancer and improve the delivery of cancer drugs by making cell membranes more permeable. Electric fields affect important pathways that cancer cells use to grow, making them more sensitive to treatments. When used with radiotherapy or chemotherapy, electric fields can make these treatments work better.Overall, the study highlights that electric fields can be a powerful addition to cancer therapy, offering multiple ways to fight the disease and improve patient outcomes.

Educate, not kill: treating cancer without triggering its defenses

This paper highlights the limitations of traditional cytotoxic therapies and the emergence of resistant cells, leading to therapy failure. Alternative therapeutic strategies, such as controlling cell dormancy, transdifferentiation therapy, normalizing the cancer microenvironment, and migrastatic therapy, are proposed as effective approaches to re-educate cancer cells towards a less malignant phenotype. These strategies aim to avoid inducing direct proliferative advantages to resistant cells, thereby delaying or preventing the development of therapy-resistant tumors. Therefore, alternative therapies are crucial for improving cancer treatment outcomes.

Alternating Electric Fields in Glioblastomas: Past, Present, and Future

Alternating electric fields is a new and noninvasive treatment method for glioblastomas, a type of deadly brain cancer. This therapy has shown promising results, including prolonged survival for patients and manageable side effects. It works by strengthening the body’s own immune response against the tumor, increasing the permeability of cell membranes and the blood-brain barrier, and disrupting the processes that repair DNA damage in cancer cells. However, despite these promising results, the acceptance of alternating electric fields in everyday clinical practice is still low. The paper calls for more studies and discussions to better understand the potential of alternating electric fields and to address any concerns that may be limiting its use in real-world settings. In simple terms, alternating electric field is a promising new treatment for a type of brain cancer, but more work needs to be done to make it a common practice in clinics. It’s an exciting development in cancer treatment, but as with all new treatments, it’s important to continue researching and understanding its full potential and limitations.

Certain types of cancers, like non-small cell lung cancer, ovarian cancer, and pancreatic cancer, are often treated with chemotherapy. However, these treatments can cause harmful side effects. There’s a need for additional therapies that can improve the effectiveness of these treatments without increasing the side effects. One such therapy is alternating electric field, which uses electric fields to disrupt the growth of cancer cells. The study suggests that using electric field therapy along with chemotherapy could potentially improve cancer treatment effectiveness without increasing side effects. This could be a promising step forward in cancer treatment.

Disruption of Cancer Cell Replication by Alternating Electric Fields

Electric fields hold potential as a therapy for cancer, particularly for blood cancers characterized by spherical suspended cells. The paper elucidates how electric fields can disrupt the cell division process, inflicting damage on cancer cells. This study aims to encourage further research and advancements in electrostatic therapy, presenting a non-invasive, cost-effective, and targeted alternative to conventional treatments.

The study delves into a captivating investigation aiming to differentiate between normal and cancer cells within liver, lung, and breast tissues. Using a set of parameters based on capacitance-voltage, researchers pinpointed unique electrical signatures for these cells. This pioneering method introduces novel prospects for recognizing and distinguishing normal and cancer cells based on their individual electrical signals. These findings offer potential advancements in diagnostic techniques, enhancing our capability to differentiate between healthy and cancerous cells across various tissue types.

Electric fields present a promising avenue for cancer therapy, particularly for blood cancers characterized by spherical suspended cells. The paper elucidates how these fields can disrupt the cell division process, inflicting harm on cancerous cells. This study aims to stimulate further exploration and advancement in electrostatic therapy, envisioning a non-invasive, cost-effective, and targeted alternative to traditional treatments.

Recent advancements of alternating electric fields therapy demonstrate potential in prolonging survival without the common side effects associated with traditional chemotherapy. This presents a promising prospect for a treatment that is both more effective and easier for patients to tolerate. As research continues, the idea of combining alternating electric field with other anti-cancer approaches emerges as a potential strategy to further boost effectiveness. Understanding these developments is essential for patients, caregivers, and the broader community, instilling hope and encouraging support for ongoing research aimed at improving outcomes for individuals with glioblastoma.

Alternating electric fields therapy stands as a promising non-invasive cancer treatment, known for its minimal side effects. This innovative therapy disrupts cancer cell division without inducing considerable systemic toxicity, presenting a hopeful avenue for a more manageable and potent approach to cancer treatment.

This research emphasizes the potential of electric fields in combating cancer cells by disrupting their functions, leading to cell death and inhibiting their growth. The efficacy of this treatment hinges on several factors, including the frequency, intensity, duration, and direction of the electric field. Furthermore, when combined with other treatments like radiotherapy or chemotherapy, electric fields often exhibit a synergistic effect, enhancing their overall effectiveness. Overall, this offers a new ray of hope, particularly for patients whose cancers show resistance to traditional treatment methods.

Alternating electric fields therapy stands as a promising non-invasive cancer treatment, known for its minimal side effects. This innovative therapy disrupts cancer cell division without inducing considerable systemic toxicity, presenting a hopeful avenue for a more manageable and potent approach to cancer treatment.

alternating electric fields therapy offer a novel strategy in combating cancer by utilizing electric fields. They possess the capability to halt the growth and spread of cancer cells while preserving normal cells from harm. Electric field are user-friendly, entail minimal side effects, and complement other treatment modalities effectively.

Electric Field based Tumor-treating fields represent a promising, novel approach in cancer treatment, employing electric fields to specifically target cancer cells while sparing normal ones. These fields are non-invasive, yield minimal side effects, and when combined with other therapies, show potential to enhance treatment outcomes.

Scientists have devised a novel approach to cancer treatment using electric fields. These fields, though extremely mild, have no detrimental impact on healthy cells; however, they possess the ability to impede the growth and division of cancer cells by influencing their internal structures. Administering these electric fields to the tumor site involves a device capable of adjusting the strength and orientation of the fields based on the cancer type. Extensive testing in both animals and humans across various cancer types has demonstrated favorable outcomes, including tumor reduction, extended survival rates, and improved patient quality of life. Notably, these electric fields have minimal side effects and synergize effectively with other treatments like surgery, chemotherapy, and radiation therapy. They possess a unique mechanism for eliminating cancer cells, distinct from conventional treatments, and can be tailored to target specific cancer cell types.

Cancer is a deadly disease that affects millions of people worldwide. Many treatments have been developed to fight cancer, but they often have serious side effects or limited effectiveness. A new technology uses electric fields to stop cancer cells from growing and spreading. Researchers have found that alternating electric field not only kill cancer cells directly, but also activate the body’s own immune system to fight cancer. This is important because the immune system can recognize and destroy cancer cells that escape other treatments. This review summarizes the current knowledge on this topic and discusses the potential benefits and challenges of combining alternating electric field with other therapies that boost the immune system.

Researchers have uncovered the effectiveness of low-intensity, intermediate-frequency alternating electric fields in halting the growth of cancer cells. This innovative method has undergone rigorous testing, proving successful in lab settings (in vitro), animal trials (in vivo), and even in a select group of human patients battling recurrent glioblastoma, a formidable brain tumor. The outcomes are nothing short of remarkable, revealing a substantial increase in the time to disease progression and overall survival rates, all while maintaining minimal side effects. This groundbreaking discovery not only offers hope for those facing challenging forms of cancer but also signifies a promising stride toward more effective and less intrusive treatment options.

Revolutionary strides in cancer treatment are unfolding through innovative methods. A recent study has unveiled a promising approach by combining alternating electric fields with the chemotherapy agent Daunorubicin, showcasing enhanced efficacy in treating blood cancer cells, particularly the non-adherent U937 cells. This cutting-edge technique selectively targets dividing cancer cells while sparing normal cells, potentially paving the way for reduced side effects in patients. It's crucial to acknowledge that these findings are preliminary, and further research is imperative to solidify their impact. As always, individuals are advised to consult their healthcare providers for personalized guidance based on their unique health circumstances. This research signifies a significant leap forward in the relentless pursuit of more effective and targeted cancer treatments.

Advancements in alternating electric fields can significantly improve the survival rates of patients with metastatic non-small cell lung cancer (mNSCLC) who have not responded to platinum-based chemotherapy. This study shows that combining alternating electric field therapy with standard-of-care treatments can lead to better outcomes compared to standard-of-care treatments alone.

The article reviews the progress of research on the mechanism of anti-tumor immune response induced by Tumor Treating Fields (TTFields). TTFields has been approved for the treatment of glioblastomas and malignant pleural mesotheliomas. It highlights that TTFields have shown promising effects as a monotherapy and in combination with chemotherapy, but the underlying mechanisms through, which TTFields exert their anticancer effects remain incompletely understood. Recent research suggests that inducing anti-tumor immune responses may be a key mechanism of the anticancer activity of TTFields, leading to several clinical trials exploring the combination of TTFields with tumor immunotherapy and achieving positive results. The article also discusses the potential mechanisms through which TTFields induce anti-tumor immune responses, including enhancing immune cell infiltration and function, inducing immunogenic cell death in tumor cells, regulating immune-related signaling pathways, and upregulating immune checkpoints in tumor cells.  Furthermore, the clinical significance of TTFields in activating and enhancing anti-tumor immune responses is highlighted, showing potential improvements in patient survival and quality of life. The combination of TTFields with immune checkpoint inhibitors has shown unprecedented therapeutic effects in clinical practice, indicating the promising clinical application prospects of TTFields. However, the document emphasizes the need for further research to clarify the molecular mechanisms of TTFields in anti-tumor therapy, potentially overcoming the problem of low sensitivity to radiotherapy and chemotherapy and enhancing the therapeutic outcomes of TTFields. Overall, the review provides a comprehensive overview of the progress and potential of TTFields in activating anti-tumor immune responses and improving clinical outcomes in cancer therapy.

This paper explores the unique electrical properties of cancer cells, shedding light on the complex network of factors that contribute to the development of cancer. It challenges the
traditional view of cancer as solely a genetic disease and emphasizes the importance of understanding the electrostatic changes in cancer cells compared to normal cells. By exploring the
effects of alterations in intracellular and extracellular pH, changes in ionic concentrations, variations in transmembrane potential, and modifications within mitochondria, the paper provides a comprehensive understanding of the electrical landscape of cells. Additionally, it discusses the potential implications of these electrical properties for novel cancer treatment modalities, such as electromagnetic field-based therapies. The research aims to pave the way for a new paradigm in understanding the role of electrical properties in health and disease, with the potential to revolutionize therapeutic interventions.

This paper demonstrate multimodal treatment approach combining electric fields and the drug repurposing strategy CUSP9v3 shows promising results in enhancing the anti-glioblastoma activity. The study provides evidence of the synergistic effects of electric fields and CUSP9v3 in inhibiting the growth and migration of glioblastoma cells. Additionally, the combination treatment was associated with the suppression of oxidative phosphorylation, a key feature of cancer cell metabolism. These findings suggest that the multimodal approach may offer a potential strategy for improving treatment outcomes for glioblastoma patients. The study also highlights the need for further research and potential transition to the clinical setting.

Electric Fields therapy has demonstrated significant improvements in overall survival (OS) and progression-free survival (PFS) when applied with adjuvant temozolomide (TMZ) compared to TMZ alone in newly diagnosed glioblastoma (ndGBM). Electric Fields therapy delivers electric fields, through scalp-placed arrays, that disrupt cellular processes critical for cancer cell viability, is CE marked for WHO grade 4 glioma, and is a recommended treatment regimen for ndGBM. Electric Fields  therapy was administered to >25,000 patients, showing no systemic toxicities and mild to moderate skin reactions being the main therapy-related adverse event. Here, the report survival and safety data from the TIGER study, the largest prospective study investigating real-world use of TTFields therapy during routine clinical care in patients with ndGBM in Germany. 

The study by Mylod et al. (2024) investigates the effects of electric fields, a non-invasive treatment using low-intensity, intermediate-frequency alternating electric fields, on natural killer (NK) cells and their ability to target glioblastoma (GBM) cells. The researchers found that electric fields, particularly at 200 kHz, significantly enhanced NK cell degranulation, a marker of cytotoxicity, against both K562 target cells and GBM cell lines without affecting NK cell viability or cytokine (IFN-γ) production.

This suggests that combining electric fields with NK cell-based immunotherapy could improve the efficacy of GBM treatments by increasing NK cell-mediated tumor cell killing. While electric fields exposure reduced the cytokine-mediated upregulation of nutrient receptors on NK cells, it did not impact mitochondrial health or granzyme B expression. These findings highlight the potential of electric fields to augment NK cell immunotherapy for GBM, warranting further investigation into the mechanisms and long-term effects of this combination treatment in more complex models.

This paper provides an extensive overview of electric fields technology, particularly its application in treating glioblastoma multiforme (GBM). Electric fields therapy is a non-invasive treatment that employs low-intensity, intermediate-frequency alternating electric fields to disrupt cancer cell division, leading to cell death while sparing normal cells. The therapy is administered through a wearable device, which patients can use at home, ensuring continuous treatment. Clinical trials have demonstrated the efficacy of electric fields in both recurrent and newly diagnosed GBM, showing comparable or superior outcomes to traditional chemotherapy with fewer side effects. Future research and clinical trials are exploring the application of electric fields in treating other types of solid tumors, with ongoing studies indicating promising synergistic effects when combined with other treatments like radiotherapy and immunotherapy. The paper underscores the importance of multidisciplinary care and continuous patient education to optimize the benefits of this innovative cancer treatment technology.

GBM is among the most lethal and challenging cancers due to their high recurrence rates and resistance to conventional therapies. Despite advancements in surgery, chemotherapy, and radiation, the prognosis for GBM remains dismal, with a median survival of only 15 months and a 5- year survival rate below 5%. This highlights the urgent need for innovative treatments. Electric field-based therapies have emerged as a promising non-invasive therapeutic approach that leverages the unique bioelectrical properties of cancer cells to disrupt their growth and proliferation. Electric fields work by applying alternating electric fields at specific frequencies and intensities, which interfere with the bioelectrical state of macromolecules and organelles within cancer cells, leading to several anti-cancer effects. These include (1) inhibition of cell mitosis by disrupting the formation of microtubule spindles, which leads to mitotic arrest and cell death; (2) disruption of genomic integrity, which increases DNA damage and inhibits DNA repair mechanisms, thus contributing to cancer cell death; (3) suppression of cell migration and invasion by altering the cellular cytoskeleton and affecting ion channel activity, thereby hindering the metastatic potential of cancer cells; (4) induction of autophagy, where abnormal mitotic events can trigger cellular self- digestion processes leading to cell death; and (5) enhancement of the anti-tumor immune response by inducing the release of damage-associated molecular patterns that activate the immune system against the tumor.

Key Findings:
1. Selective Apoptosis:

  • The study demonstrated that Positive Electrostatic Charges (PECs) could selectively induce apoptosis in breast cancer cell lines, including MCF-7 (hormone receptor-positive breast cancer) and MDA-MB-468 (triple-negative breast cancer) cells. The therapy showed significant reductions in cell viability, while normal breast epithelial cells (MCF-10A) were not adversely affected, highlighting the selectivity of PECs.


2. Mechanism of Action:

 

 

  • PECs were found to disrupt the cytoskeleton and critical metabolic pathways in cancer cells, leading to apoptosis. This effect was particularly noted in MCF-7 cells, where PECs caused disruptions in the cell cycle and induced apoptosis through the mitochondrial pathway, evidenced by changes in the expression of apoptosis-related proteins. 

  • MCF-10A Control: The lack of similar disruptions in MCF-10A cells underscores the specificity of PECs for malignant cells, making it a promising therapeutic approach with minimal risk to normal tissues.


3. In Vivo Validation:

  • The study extended these findings to animal models, where tumors derived from MCF-7 cells were treated with PECs. Significant tumor reduction was observed in the treated animals, with no adverse effects on surrounding normal tissues, indicating that PECs could effectively target tumors without collateral damage.


Clinical Impact:
1. Non-Invasive Cancer Treatment:

  • PEC therapy offers a promising non-invasive treatment option for breast cancer, particularly for patients with tumors that are resistant to standard therapies. The ability to selectively target cancer cells like MCF-7 while sparing normal cells such as MCF-10A could lead to fewer side effects and better overall patient outcomes.


2. Potential for Metastatic and Hormone-Responsive Breast Cancer:

  • Given the effectiveness of PECs in targeting both MCF-7 (hormone receptor-positive) and MDA-MB-468 (triple-negative) breast cancer cells, this therapy could be applied to a broad spectrum of breast cancer subtypes, including those that are metastatic or hormone-responsive.
     

3. Foundation for Human Trials:

  • The strong preclinical evidence, particularly the selectivity demonstrated in both cancerous (MCF-7, MDA-MB-468) and non-cancerous (MCF-10A) cells, supports the initiation of human trials. Successful human trials could lead to PEC being integrated into treatment protocols for various types of breast cancer.

Key Findings:
1. Effectiveness Against Circulating Tumor Cells:

  • The study demonstrated that PECs could effectively deactivate circulating tumor cells (CTCs) in the bloodstream. The efficacy of PECs was evaluated using both in vitro and in vivo models, including human metastatic breast cancer cell lines (MDA- MB-231) and mouse mammary carcinoma cell lines (4T1).

  • CTC Deactivation: PECs treatment significantly reduced the viability and metastatic potential of CTCs. Treated cells showed diminished spheroid formation ability, reduced invasive behavior, and lower metastatic potential in subsequent in vivo tests, indicating that PECs effectively neutralize the cells responsible for spreading cancer.


2. In Vivo Efficacy in Mouse Models:

  • 4T1 Cells: In BALB/c mice injected with 4T1 cells (a highly metastatic breast cancer cell line), PECs significantly inhibited the development and progression of primary tumors. Notably, only one out of ten mice developed a tumor when treated with PECs, compared to nine out of ten in the untreated control group.

  • Metastasis Reduction: PECs also led to a substantial reduction in metastatic spread, particularly to the lungs. Mice treated with PECs had significantly fewer lung nodules compared to controls, demonstrating the treatment’s potential to prevent metastasis.

3. Impact on Normal Cells:

  • White Blood Cells (WBCs): The study included an assessment of normal WBCs to evaluate the safety of PECs. Flow cytometry analysis revealed that less than 9% of WBCs were affected by PECs treatment, indicating a minimal impact on these normal cells.

  • Histological Analysis: Further histological examinations of tissues from the treated mice showed no significant damage to normal organs or tissues, confirming the selectivity and safety of PECs.

4. Mechanistic Insights:

  • Selective Targeting: The study provided mechanistic insights into how PECs selectively targets malignant cells. PECs disrupt critical cellular processes and structures in CTCs, leading to a decrease in cell proliferation and invasion while sparing normal cells like WBCs. This selective action is crucial for its application in clinical settings where minimizing harm to healthy tissue is paramount.


Clinical Impact:
1. Novel Strategy for Metastasis Prevention:

  • PECs present a new, non-invasive method for preventing metastasis by targeting and deactivating CTCs in the bloodstream before they can establish secondary tumors. This approach could be particularly valuable for patients with aggressive cancers, where early intervention can prevent the spread of the disease and improve survival outcomes.


2. Improved Survival Outcomes:

  • By effectively reducing the number of viable CTCs and their ability to form new metastatic sites, PECs has the potential to significantly improve long-term survival rates in patients at high risk of metastasis. This is especially important in cancers like triple-negative breast cancer, where metastasis often leads to poor prognosis.


3. Selective Targeting with Minimal Side Effects:

  • The study’s findings that PECs selectively target cancer cells while sparing normal cells, such as WBCs, underscore its potential as a safe and targeted therapy. This selectivity minimizes the risk of side effects, making PECs a promising candidate for clinical use, particularly in patients who are not suitable for more aggressive treatments.

4. Foundation for Clinical Trials:

  • The promising preclinical results from this study provide a strong foundation for advancing PECs to clinical trials in human patients. If similar safety and efficacy are observed in human studies, PECs could become an integral part of treatment protocols for managing metastatic cancer, particularly in breast cancer patients.


5. Broad Applicability:

  • While the study primarily focused on breast cancer models, the principles behind PECs suggest it could be applied to a wide range of cancers where metastasis is a major concern. This broad applicability makes PECs a potentially transformative tool in oncology, offering a new line of defense against the spread of cancer.

Key Findings:
1. Patient Demographics and Study Design:

  • This human pilot study involved 41 patients with late-stage metastatic cancer, all with solid tumors that were unresponsive to conventional therapies such as chemotherapy and radiotherapy. These patients were considered for Positive Electrostatic Charge Therapy (PECT) as a last resort.


2. Tumor Response:

  • Significant Tumor Reduction: Over 80% of the patients exhibited a measurable reduction in tumor size, with some tumors shrinking by more than 50%. This significant reduction occurred without disease progression during the treatment period.

  • Stabilization of Disease: For patients who did not experience significant shrinkage, the disease was stabilized, with no further tumor growth observed.


3. Symptom Relief and Quality of Life:

  • Improvement in Symptoms: Patients reported relief from various cancer-related symptoms, such as pain and fatigue, leading to an overall improvement in quality of life. Some patients also experienced improved mobility and daily functioning.


4. Mechanism of Action:

  • The therapy is believed to work by disrupting cancer cell metabolism and inducing apoptosis through the modulation of the KRAS signaling pathway and increasing the Bax/Bcl2 ratio, favoring cell death.

  • No Damage to Normal Tissues: The treatment did not result in adverse effects on surrounding normal tissues, avoiding common side effects like skin burns or inflammation.

5. Safety Profile:

  • No Major Adverse Events: The treatment was well-tolerated by all patients, with no reports of severe side effects commonly associated with traditional cancer therapies. The absence of side effects like nausea, vomiting, or hair loss highlights the therapy's safety.


Clinical Impact:
1. Potential as a Last-Resort Therapy:

  • For patients with late-stage metastatic cancer who have no other treatment options, PECT offers a potentially effective and safe alternative, extending life and improving quality of life.


2. Non-Invasive and Safe Alternative:

  • The non-invasive nature of PECT, coupled with its minimal side effects, positions it as a safer alternative to more toxic treatments like chemotherapy and radiotherapy.

  • This could be especially beneficial for patients who are not candidates for aggressive treatments.


3. Foundation for Larger Clinical Trials:

  • This pilot study provides a strong basis for larger, more comprehensive clinical trials. If similar results are replicated in larger populations, PECT could gain regulatory approval and become a standard treatment option for advanced cancers.


4. Broad Application Potential:

  • While this study focused on late-stage metastatic cancers, the underlying mechanisms suggest that PECT could be applicable to a variety of cancer types, potentially expanding its use across different patient groups.

bottom of page