Pulsed Electromagnetic Fields and Tumor-Targeted Radiofrequencies in Cancer Therapy: A Review of Experimental and Clinical Evidence
Abstract
Pulsed
electromagnetic fields (PEMF) and amplitude-modulated radiofrequency electromagnetic
fields (AM-RF EMF) are emerging as non-invasive adjuncts in cancer therapy.
Initially approved for bone healing and pain management, PEMF has shown
potential in inhibiting tumor cell proliferation, inducing apoptosis, and
impairing angiogenesis in various preclinical models. This review synthesizes
findings from studies involving low- and high-intensity PEMF, tumor-treating
alternating electric fields, and tumor-specific AM-RF EMF across cancer cell
lines, animal models, and early-phase human trials. While results are
promising, therapeutic responses remain highly context-dependent and
mechanistically heterogeneous. Advances in structural biology and cancer
genomics offer new opportunities to rationally design field parameters
targeting specific molecular pathways. We propose a framework for estimating
the electromagnetic field strength needed to disrupt key protein-ligand
interactions and highlight pathways for future investigation. Rigorous clinical
trials and optimized protocols will be essential to fully integrate PEMF into
precision oncology.
Introduction
Pulsed
Electromagnetic Field (PEMF) therapy is an FDA-approved, non-invasive modality
widely used in orthopedics to support bone healing in conditions such as
nonunion fractures, spinal fusions, and osteotomies. Devices like Orthofix’s
Physio-Stim and Biomet’s EBI Bone Healing System deliver time-varying
electromagnetic fields to stimulate cellular activity and promote osteogenesis
and angiogenesis. These effects are mediated through mechanisms including
modulation of ion binding, upregulation of growth factors (e.g., BMP-2, TGF-β),
and increased expression of osteogenic markers (Bassett et al., 1974; Aaron et
al., 2004).
Beyond
orthopedics, PEMF is gaining attention as a complementary therapy for chronic
pain, particularly musculoskeletal conditions such as osteoarthritis,
fibromyalgia, and chronic low back pain. Although not FDA-approved for general
pain relief, over-the-counter devices (e.g., Oska Pulse, BEMER) are marketed
under Class I or II wellness exemptions. Studies suggest PEMF may reduce pain
and improve function by modulating inflammation, nociceptive signaling, and
microcirculation (Foley-Nolan et al., 1990; Thomas et al., 2007).
Emerging
evidence now points to a potential role for PEMF in cancer therapy. Preclinical
studies demonstrate that specific PEMF exposures can inhibit tumor cell
proliferation, induce apoptosis, and impair angiogenesis in models of breast,
lung, and brain cancers. Animal studies support these findings (Vadala et al.,
2016), and early clinical trials suggest PEMF may improve quality of life and
even exert direct antitumor effects in certain contexts (Zimmermann et al.,
2012).
While
clinical data are still limited, recent reviews emphasize the need for rigorous
trials to optimize treatment protocols and clarify mechanisms (Xu et al., 2022,
Egg & Kietzmann, 2025). This review examines key findings from cellular and
animal studies, as well as preliminary human trials, tracing the evolution of
this field from early research on alternating fields and modulated
radiofrequencies to current interest in PEMF for cancer treatment.
Low and intermediate
frequency alternating electromagnetic fields
Alternating
electric fields in the intermediate frequency range (100–300 kHz) at field
strengths of 1–2.5 V/cm (corresponding to magnetic fields of 0.44–1.1 μT) have
been shown to suppress the proliferation of various rodent and human tumor cell
lines—including Patricia C, U-118, U-87, H-1299, MDA231, PC3, B16F1, F-98, C-6,
RG2, and CT-26—as well as malignant tumors in animal models. This inhibitory
effect is both frequency- and intensity-dependent and is selective for actively
dividing cells; non-proliferating (quiescent) cells remain unaffected. The mechanism
of action of these tumor treating fields (TTFields) involves disruption of
mitotic spindle formation, resulting in mitotic arrest and cell death (Kirson
et al., 2004). These findings have been extended to additional cell lines
(e.g., MDA-MB-231 and H1299) and tumor models (e.g., intradermal B16F1 melanoma
and intracranial F-98 glioma). In a pilot clinical study of 10 glioblastoma
patients, TTFields therapy more than doubled both median progression-free
survival and overall survival compared to historical controls (Kirson et al.,
2007).
In contrast,
exposure of Caco-2 human colon adenocarcinoma cells to low-frequency (50 Hz),
higher-intensity (1 mT) magnetic fields promoted cell growth rather than
inhibiting it. This stimulation was time-dependent and accompanied by increased
protein oxidation and elevated intracellular reactive oxygen species (ROS).
These changes coincided with increased intracellular calcium levels and global
activation of the 20S proteasome, along with a reduction in the pro-apoptotic
protein p27 (Eleuteri et al., 2009).
Other
studies have reported no significant effects of 1 mT, 60 Hz electromagnetic
fields on non-cancerous immortalized cell lines such as Jurkat (human T
lymphocytes) and NIH3T3 (mouse embryonic fibroblasts). However, under the same
conditions, both MCF-7 (human breast cancer) and MCF-10A (non-tumorigenic
breast epithelial) cells exhibited significant reductions in cell number,
viability, and DNA synthesis. These effects were attributed to cell cycle delay
and induction of the pro-apoptotic gene PMAIP in a context-dependent manner
(Lee et al., 2015).
Overall, the
biological effects of low- and intermediate-frequency EMFs on cancer cells are
complex and highly context-dependent. Responses vary by cell type,
proliferative status, field parameters (frequency and intensity), and exposure
duration, underscoring the importance of precise characterization in
therapeutic and experimental applications.
Amplitude
modulated radiofrequency electromagnetic fields
In 2009,
Barbault et al. reported that cancer patients exhibited biofeedback responses
to tumor-specific frequencies of amplitude-modulated (AM) radiofrequency
electromagnetic fields (RF-EMF). These modulation frequencies, ranging from 0.1
Hz to 114 kHz, were specific to the type of tumor, while the carrier frequency
was a fixed 27.12 MHz. The RF signal was generated at 100 mW into a 50-ohm
load. In a limited compassionate-use clinical trial involving 28 patients with
various cancer types, RF-EMF treatment was delivered intrabuccally for 60
minutes, three times daily, and continued until disease progression or death.
No
significant side effects were reported. Of the 13 patients eligible for
response evaluation, one breast cancer patient achieved a complete response,
and another showed a partial response. Four additional patients (with thyroid,
lung, pancreatic cancers, and leiomyosarcoma) exhibited stable disease. The
authors concluded that the observed clinical responses were more likely due to
systemic physiological effects rather than direct cytotoxic action, given the
low field strength and the anatomical distance between the intrabuccal
application site and the tumor sites. Estimated field strengths within 1 mm of
the emitter were approximately 30 V/cm (electric) and 13 μT (magnetic).
A subsequent
open-label phase I/II clinical trial involving 41 patients with hepatocellular
carcinoma (HCC) confirmed the earlier findings. Using the same protocol of
tumor-specific AM RF-EMF delivery, 28 patients were evaluable for response: 4
demonstrated partial responses, 16 had stable disease, and 8 showed disease
progression. These preliminary outcomes were considered promising and formed
the rationale for pursuing larger, randomized clinical trials (Costa et al.,
2011).
Follow-up
mechanistic studies investigated the effects of tumor-specific modulation
frequencies on HCC (HepG2, Huh7) and breast cancer (MCF-7) cell lines. Direct
in vitro exposure resulted in significant growth inhibition of malignant cells,
whereas non-malignant counterparts—THLE-2 hepatocytes and MCF-10A breast
epithelial cells—were unaffected. Growth suppression in HCC cells was
accompanied by downregulation of the chemokine-related genes XCL2 and PLP2,
as well as disruption of mitotic spindle architecture. Notably, reduced
expression of XCL2 and PLP2 has been associated with improved
prognosis in cancer patients (Zimmerman et al., 2012).
Low intensity
PEMF
The
biological effects of pulsed electromagnetic fields (PEMF) on tumor cell growth
have been recognized for over two decades. Early studies demonstrated that PEMF
exposure at a magnetic field intensity of 1.5 mT and pulse frequencies of 1 or
25 Hz enhanced the cytotoxicity of chemotherapeutic agents—vincristine,
mitomycin, and cisplatin—against multidrug-resistant HCA-2/1cch human colon
adenocarcinoma cells in vitro (Ruiz-Gómez, M.J., 2002). At the same field
intensity but a higher pulse frequency (75 Hz), PEMF upregulated A3 adenosine
receptor (A3AR) expression in neural tumor cell lines, including PC12 (rat adrenal
pheochromocytoma) and U87MG (human glioblastoma). This upregulation was
associated with inhibition of NF-κB signaling and induction of p53, ultimately
leading to suppressed proliferation, increased lactate dehydrogenase (LDH)
release, and elevated caspase-3 activity—markers of cytotoxicity and apoptosis,
respectively (Vincenzi, F., 2012). Similarly, growth inhibition and apoptotic
induction were observed in the SKOV3 human ovarian cancer cell line following
PEMF exposure at 1 mT with pulse frequencies ranging from 8 to 64 Hz (Wang et
al., 2012).
More recent
studies have extended these findings to other tumor types. In vitro and in vivo
experiments involving human breast cancer MCF-7 cells and human lung
adenocarcinoma A549 cells revealed that low-intensity PEMF (0.68 and 1.19 mT)
applied at higher pulse frequencies (3.846 and 40.85 kHz, respectively)
significantly inhibited tumor growth (Chen et al., 2022). This effect was
attributed to increased apoptotic activity, evidenced by elevated caspase-3/7
expression and greater annexin V staining, as well as an accumulation of cells
in the G0 phase of the cell cycle. Gene expression analysis further
indicated activation of pathways associated with DNA damage, cell cycle arrest,
and growth suppression.
Notably, the
systemic impact of PEMF has also been demonstrated in humans. In a recent
double-blind, randomized clinical trial involving healthy female volunteers,
participants were exposed to PEMF at 1 mT intensity and 50 Hz pulse frequency
(Iversen et al., 2025). Sera collected from treated individuals exhibited
significant anti-cancer properties up to one month post-exposure, reducing
breast cancer cell proliferation, migration, and invasiveness in vitro. These
effects correlated with a reduction in epithelial-mesenchymal transition (EMT)
markers, suggesting a systemic modulation of anti-tumor signaling pathways.
Higher
intensity PEMF exposure
For the
purposes of this review, high-intensity pulsed electromagnetic fields (PEMF)
are defined as those with magnetic field strengths ranging from 2 to 400 mT,
remaining within the public exposure limits recommended by the International
Commission on Non-Ionizing Radiation Protection (ICNIRP) (Yamaguchi-Sakeno et
al., 2011). The specific pulse frequencies and exposure durations varied across
the studies discussed below.
Over the
past two decades, a growing body of research involving both tumor cell lines
and animal models has demonstrated that PEMF—either as a stand-alone treatment
or in combination with chemotherapy or gamma irradiation—can exert
antiproliferative and antiangiogenic effects. These studies typically used
field intensities between 2 and 20 mT, pulse frequencies from 8 to 120 Hz, and
diverse exposure regimens. Tumor types studied included breast, bladder, liver,
hematopoietic cancers, osteosarcoma, and fibrosarcoma.
Breast
cancer cell lines, particularly MCF-7, have shown notable sensitivity to PEMF.
Exposure to PEMF at 3 mT and 20 Hz for 60 minutes daily over three days
significantly inhibited proliferation (Crocetti et al., 2013). A separate
protocol using full-square wave PEMF at 11 mT and 8 Hz, applied for 30 minutes
twice daily over five days, yielded similar antiproliferative outcomes in both
MCF-7 and MDA-MB-231 breast cancer cells (Pantelis et al., 2024). These effects
were mechanistically linked to DNA damage, apoptosis, and the induction of
cellular senescence markers. Importantly, these effects appeared to be
selective for malignant cells; normal epithelial and fibroblast cells remained
unaffected under the same treatment conditions.
The
potential for PEMF to enhance chemotherapy has also been explored. In MCF-7
cells, PEMF exposure significantly potentiated the cytotoxic effects of
doxorubicin (Woo & Kim, 2024) and etoposide (Woo et al., 2022). Both agents
inhibit cell proliferation through topoisomerase II inhibition and reactive
oxygen species (ROS) generation, and these pathways appeared to be further
activated in the presence of PEMF.
Findings
from in vitro studies have been validated in vivo. For example, PEMF exposure
inhibited the growth and vascularization of 16/C murine mammary adenocarcinoma
tumors implanted in syngeneic C3H/HeJ mice. Treatment consisted of 10-minute
daily exposures for 12 days using a 120 Hz pulse frequency and field
intensities up to 20 mT (Williams et al., 2001). Further studies confirmed that
tumor inhibition was dependent on increasing magnetic field intensity rather
than increased exposure time at a fixed intensity (Cameron et al., 2014). PEMF
also enhanced the effects of gamma irradiation and the chemotherapeutic agent
bleomycin in mouse models bearing human MDA-MB-231 breast cancer xenografts
(Cameron et al., 2005) and triple-negative breast cancer (TNBC) MX-1 xenografts
in SCID mice (Berg et al., 2010).
Beyond
breast cancer, PEMF has demonstrated antitumor activity in models of bladder
cancer (Sanberg et al., 2025), hematologic malignancies (Radeva & Berg,
2004; Berg et al., 2010), osteosarcoma (Muramatsu et al., 2017), and
fibrosarcoma (Omote et al., 1990). Of particular interest is the reported
synergy between PEMF and molecularly targeted therapies: in BCR/ABL(+)
leukemia-derived TCC-S cells, PEMF enhanced the efficacy of the tyrosine kinase
inhibitor imatinib (Yamaguchi-Sakeno et al., 2011).
Future
directions
The growing
body of evidence supports the continued development of pulsed electromagnetic
fields (PEMF) as a complementary modality in cancer therapeutics. At present,
PEMF demonstrates its greatest efficacy when combined with conventional
treatments, such as cytotoxic chemotherapy or ionizing radiation. Several novel
laboratory protocols have yielded promising results and merit translation into
large-scale, double-blind clinical trials to evaluate therapeutic utility and
safety in a controlled setting. However, current findings also highlight
important areas for further refinement and optimization.
To date, no
PEMF or EMF protocol has consistently achieved irreversible tumor regression as
a stand-alone intervention. Moreover, the heterogeneity in field
parameters—such as frequency, waveform, intensity, and exposure duration—across
studies has impeded the establishment of a unified mechanism of action.
Inhibition of cancer cell growth by PEMF appears to involve multiple, and
potentially interacting, cellular processes, including alterations in membrane
potential, disruption of mitochondrial function, interference with mitotic
spindle formation, modulation of growth signaling pathways, increased
generation of reactive oxygen species (ROS), and induction of apoptosis. While
the preferential sensitivity of malignant cells compared to normal cells is
encouraging, the underlying basis of this selectivity remains incompletely
understood and warrants further mechanistic investigation.
Reliance on
empirical trial-and-error testing may be inefficient and limiting, especially
given current advances in cancer genomics and the characterization of key
oncogenic driver mutations (Kinnersley et al., 2024). Moving forward, it would
be advantageous to rationally design PEMF protocols that specifically disrupt
molecular pathways activated by such driver genes. This approach could yield
targeted, mechanism-informed applications of PEMF with improved therapeutic
indices. Table 1 summarizes major classes of cancer-relevant signaling pathways
and representative driver genes identified through high-throughput genome
sequencing.
Many of the
downstream effectors of cancer-associated gene products—such as enzymes,
receptors, transcription factors, DNA-binding proteins, and scaffolding
proteins—have had their structures experimentally determined and deposited in
the Protein Data Bank (PDB), or accurately predicted by AlphaFold.
This structural information provides a powerful foundation for estimating the
electromagnetic field (EMF) intensities required to perturb or disrupt their
functional interactions. In particular, these insights open the door to the
rational design of PEMF protocols targeting specific molecular interactions
central to tumor growth and survival.
Functionally
disrupting a protein's activity via EMF can be conceptualized as interfering
with its interactions with a substrate, ligand, DNA target, or binding partner.
This disruption can occur if the energy imparted by the EMF is sufficient to
overcome the standard free energy of binding (ΔG⁰), or alternatively, the free
energy required to partially unfold one or both interacting molecules (ΔGᵤ).
For binding
interference, the minimum energy supplied by the EMF must match or exceed the
standard free energy of binding. On a per-molecule basis, this requirement can
be written as:
Eemf
= (ΔG0/N) [1]
where N is
the Avogadro number (6.022 x 1023 mol-1) since ΔG0
values are often given per mole.
The energy
provided by an electric field (E) to a dipole or charge depends on the
interaction between the field and the molecule dipole moment (μ) or charge (q). For a dipole in an
electric field, the potential energy (U) is the vectorial product of the
electric field (E) and the dipole moment (μ), and maximum energy occurs when
the dipole is aligned with the field and
Um
= μE [2]
where μ is
the magnitude of the dipole moment in Debyes or C.m, and E the electric field
strength in V/m.
Since the
energy required by the field to disrupt binding must at least be equal to the
free binding energy,
μE = ΔG0/N [3]
Solving for
E,
E = ΔG0/(Nμ) [4]
In practice,
ΔG0 could be derived from the equilibrium binding constant Keq
using,
ΔG0
= -RTlnKeq [5]
where R is
the universal gas constant ( ≈ 8.314 J⋅mol⁻¹⋅K⁻¹) and T the
temperature in Kelvin (K).
The corresponding
magnetic field strength could be derived from,
B = E/c [6]
a consequence of Maxwell’s equations and where
c is the speed of light (≈ 3 x 108 m/s).
Table 2 shows
the calculated magnetic field intensities required for disrupting the functions
of selected therapeutic targets in cancer pathways. These calculated values
should be regarded as preliminary approximations, given several limitations in
the underlying dipole moment estimations. Notably, solvent effects and
electrostatic contributions from bound ligands were not included, even though
they may significantly alter the net dipole moment of the complex. Despite
these limitations, the estimates can serve as a useful starting point for the
design of more targeted and effective pulsed electromagnetic field (PEMF)
protocols—particularly with respect to field strength, pulse frequency, and
exposure duration.
As discussed
earlier, a complementary approach to estimating the required magnetic field
intensity involves the use of the free energy of unfolding (ΔGu) of
the protein or protein-ligand complex. However, accurate ΔGu values
typically require differential scanning calorimetry (DSC) data, which remain
sparse in the literature for many of the cancer-associated targets included in
this analysis.
Table 2 indicates
that, in most cases, the magnetic field strengths needed to interfere with key
oncogenic pathways fall within the operational range of FDA-approved PEMF
devices used for specific clinical indications. Furthermore, some PEMF systems
marketed for general wellness are capable of producing high peak magnetic field
intensities—up to 100 mT—though often at lower frequencies. Clinical experience
in areas such as bone regeneration and pain control suggests that efficacy is
not solely determined by maximum field strength. Rather, an optimized
combination of intensity, frequency, pulse width, and duty cycle is likely
required for therapeutic benefit.
Future
advances in PEMF technology—guided by personalized cancer genomic data and
supported by rigorous clinical trials—may help establish electromagnetic field
modulation as a viable adjunct or alternative in cancer therapy. Such progress
will be essential to gaining broader acceptance within the medical community
and among the general public.
References
Aaron, R.
K., Ciombor, D. M., & Simon, B. J. (2004). Treatment of nonunions with
electric and electromagnetic fields. Clinical Orthopaedics and Related
Research, (419), 21–29.
Bassett, C.
A. L., Pawluk, R. J., & Pilla, A. A. (1974). Augmentation of bone repair by
inductively coupled electromagnetic fields. Science, 184(4136), 575–577.
Foley-Nolan,
D., Barry, C., Coughlan, R. J., et al. (1990). Pulsed high-frequency (27
MHz) electromagnetic therapy for persistent neck pain: a double-blind,
placebo-controlled study of 20 patients. Orthopedics, 13(4), 445–451.
Thomas, A.
W., Graham, K., Prato, F. S., et al. (2007). A randomized, double-blind,
placebo-controlled clinical trial using a low-frequency magnetic field in the
treatment of musculoskeletal chronic pain. Pain Research & Management,
12(4), 249–258.
Vadalà, M.,
Morales-Medina, J. C., Vallelunga, A., et al. (2016). Mechanisms and
therapeutic effectiveness of pulsed electromagnetic field therapy in oncology. Cancer
Medicine, 5(11):3128–3139.
Zimmermann,
J. W., Pennison, M. J., Brezovich, I., et al. (2012). Cancer cell
proliferation is inhibited by specific modulation frequencies. British
Journal of Cancer, 106(2), 307–313.
Xu, W., Xie,
X. Wu, H. et al. (2022). Pulsed electromagnetic therapy in cancer
treatment: progress and outlook. View 3, 20220029.
Egg, M.
& Kietzmann, T. (2025). Little strokes fell big oaks: The use of weak
magnetic fields and reactive oxygen species to fight cancer. Redox Biology
79, 103483.
Kirson, E.
D., Gurvich, Z., Schneiderman, R. et al.(2004). Disruption of Cancer Cell
Replication by Alternating Electric Fields. Cancer Research 64,
3288–3295.
Kirson, E.
D., Dbaly, V., Tovarys, F. et al. (2007). Alternating electric fields
arrest cell proliferation in animal tumor models and human brain tumors. Proceedings
of the National Academy of Sciences 104, 10152–10157.
Eleuteri, A.
M., Amici, M., Bonfili, L. et al. (2009). 50 Hz extremely low frequency electromagnetic
fields (ELF-EMF) enhance protein carbonyl groups content in cancer cells: effects
on proteasomal systems. Journal of Biomedicine and Biotechnology Article
ID 834239.
Lee, H.C.,
Hong, M-N., Jung, S.H., et al. (2015). Effect of extremely low frequency
magnetic fields on cell proliferation and gene expression. Bioelectromagnetics,
36: 506-516.
Barbault, A.,
Costa, F. P., Bottger, B. et al. (2009). Amplitude-modulated
electromagnetic fields for the treatment of cancer: Discovery of tumor-specific
frequencies and assessment of a novel therapeutic approach. Journal of
Experimental & Clinical Cancer Research 28, 51.
Costa, F.
P., de Oliveira, A. C., Meirelles, R. et al. (2011).Treatment of
advanced hepatocellular carcinoma with very low levels of amplitude-modulated
electromagnetic fields. British Journal of Cancer 105,640–648.
Zimmerman,
J.W., Pennison, M.J., Brezovich, I. et al. (2012). Cancer cell
proliferation is inhibited by specific modulation frequencies. British Journal
of Cancer 106, 307–313.
Ruiz-Gómez,
M.J., de la Peña, L., Prieto-Barcia, M.I., et al. (2002). Influence of 1
and 25 Hz, 1.5 mT magnetic fields on antitumor drug potency in a human
adenocarcinoma cell line. Bioelectromagnetics 23, 578-525. https://doi.org/10.1002/bem.10054
Vincenzi, F.,
Targa, M., Corciulo, C., et al. (2012). The Anti-Tumor Effect of A3
Adenosine Receptors Is Potentiated by Pulsed Electromagnetic Fields in Cultured
Neural Cancer Cells. PloS ONE 7, e39317. doi:10.1371/journal.pone.0039317
Wang, Q., Wu, W., Chen, X., et al. (2012). Effect of pulsed
electromagnetic field with different frequencies on the proliferation,
apoptosis, and migration of human ovarian cancer cells. Sheng wu yi xue Gong
Cheng xue za zhi = Journal of Biomedical Engineering 29, 291-295. (Language:chinese)
Chen, M.Y., Li, J., Zhang, N., et al. (2022). In Vitro and in Vivo Study of
the Effect of Osteogenic Pulsed Electromagnetic Fields on Breast and Lung
Cancer Cells. Technology in Cancer Research & Treatment 21.
doi:10.1177/15330338221124658
Iversen,
J.N., Tai, Y.K., Jasmine, Yap, J.L.Y., et al. (2025). One Month of Brief
Weekly Magnetic Field Therapy Enhances the Anticancer Potential of Female Human
Sera: Randomized Double-Blind Pilot Study. Cells 14, 331.
https://doi.org/10.3390/ cells14050331
Yamaguchi-Sakino,
S., Sakino, M. & Ueno, S. (2011). Biological Effects of Electromagnetic
Fields and Recently Updated Safety Guidelines for Strong Magnetic Fields. Magn.
Reson. Med. Sci. 10, 1-10. https://doi.org/10.2463/mrms.10.1
Pantelis, P., Theocharous, G., Veroutis, D., et al.
(2024). Pulsed
Electromagnetic Fields (PEMFs) Trigger Cell Death and Senescence in Cancer
Cells. Int. J. Mol. Sci. 25, 2473. https://doi.org/ 10.3390/ijms25052473
Woo, S-H.
& Kim, Y.S. (2024). Pulsed Electromagnetic Field Enhances
Doxorubicin-induced Reduction in the Viability of MCF-7 Breast Cancer Cells. Korean
J Clin Lab Sci. 56, 73-84.
Woo, S-H., Kim,
B., Kim, S.H., et al. (2022). Pulsed electromagnetic field potentiates
etoposide-induced MCF-7 cell death. BMB Rep. 55, 148-153. https://doi.org/10.5483/BMBRep.2022.55.3.119
Williams,
C.D., Markov, M.S., Hardman, W.E. et al. (2001). Therapeutic
Electromagnetic Field Effects on Angiogenesis and Tumor Growth. Anticancer
Research 21, 3887-3892.
Cameron,
I.L., Markov, M. and Hardman, W.E. (2014). Optimization of a therapeutic
electromagnetic field (EMF) to retard breast cancer tumor growth and
vascularity. Cancer Cell International 14,125. http://www.cancerci.com/content/14/1/125
Cameron,
I.L., Sun, L-Z., Short, N. et al. (2005). Therapeutic Electromagnetic
Field (TEMF) and gamma irradiation on human breast cancer xenograft growth,
angiogenesis and metastasis. Cancer Cell International 5, 23. doi:10.1186/1475-2867-5-23
Berg, H.,
Gunther, B., Hilger, I. et al. (2010). Bioelectromagnetic Field Effects
on Cancer Cells and Mice Tumors. Electromagnetic Biology and Medicine
29, 1–12. doi: 10.3109/15368371003776725
Sanberg, M.,
Whitman, W. Bissette, R. et al. (2025). Pulsed Electromagnetic Field
Therapy Alters the Genomic Profile of Bladder Cancer Cell Line HT-1197. J.
Pers. Med. 15, 143. https://doi.org/10.3390/ jpm15040143
Sanberg, M.,
Whitman, W., Ross, C.. et al. (2025). Pulsed electromagnetic field
therapy's effect on bladder cancer cell line HT-1376. Journal of Clinical
Oncology 42 (4, suppl.). https://doi.org/10.1200/JCO.2024.42.4_suppl.666
Radeva, M.
and Berg, H. (2004). Differences in lethality between cancer cells and human
lymphocytes caused by LF-electromagnetic fields. Bioelectromagnetics 25,
503-507. https://doi.org/10.1002/bem.20023
Muramatsu, Y., Matsui, T., Deie, M. et al. (2017). Pulsed Electromagnetic Field
Stimulation Promotes Anti-cell Proliferative Activity in Doxorubicin-treated
Mouse Osteosarcoma Cells. In vivo 31, 61-68. doi:10.21873/invivo.11026
Omote, Y.,
Hosokawa, M., Komatsumoto, M. et al. (1990). Treatment of experimental
tumors with a combination of pulsing electromagnetic field and antitumor drug. Jpn.
J. Cancer Res. 81, 956-961.
Kinnersley,
B., Sud, A., Everall, A. et al. (2024). Analysis of 10,478
cancer genomes identifies candidate driver genes and opportunities for
precision oncology. Nat Genet 56, 1868–1877. https://doi.org/10.1038/s41588-024-01785-9
John, J.
Rensland, H., Schlichting, I. et al. (1993). Kinetic and Structural
Analysis of the Mg2+-binding Site of the Guanine Nucleotide-binding Protein p21H-ras.
J. Biol. Chem. 268, 923-929.
Tran, T.H.,
Chan, A.H., Young, L.C. et al. (2021). KRAS interaction with
RAF1 RAS-binding domain and cysteine-rich domain provides insights into
RAS-mediated RAF activation. Nat Commun 12, 1176 (2021). https://doi.org/10.1038/s41467-021-21422-x
Knight, Z.A.
and Shokat, K.M. (2005). Features of Selective Kinase Inhibitors. Chemistry
& Biology 12, 621 – 637. DOI: 10.1016/j.chembiol.2005.04.011
Bossi, R.T.,
Saccardo, M.B., Ardini, E. et al.
(2010). Crystal Structures of Anaplastic Lymphoma Kinase in Complex with ATP competitive
Inhibitors . Biochemistry 49, 6813–6825. DOI:
10.1021/bi1005514
Petrosino, M., Novak, L., Pasquo, A. et al. (2023).The complex impact of
cancer-related missense mutations on the stability and on the biophysical and
biochemical properties of MAPK1 and MAPK3 somatic variants. Hum
Genomics 17, 95. https://doi.org/10.1186/s40246-023-00544-x
Maheshwari,
S., Miller, M.S., O’Meally, R., et al. (2017). Kinetic and structural
analyses reveal residues in phosphoinositide 3-kinase α that are critical for
catalysis and substrate recognition. J. Biol. Chem. 292, 13541 – 13550.
DOI: 10.1074/jbc.M116.772426
Thorsell
A-G., Ekblad, T., Karlberg, T., et al. (2016). Structural Basis for
Potency and Promiscuity in Poly(ADP-ribose) Polymerase (PARP) and Tankyrase
Inhibitors. J Med Chem. Dec 21;60(4):1262–1271. doi: 10.1021/acs.jmedchem.6b00990
Table 1. Selected
Cancer Driver Genes
_________________________________________________________________________________________
Classical
Tumor Suppressors & Oncogenes
Gene |
Mutation
Frequency |
Primary
Pathway |
Major
Cancer Types |
Therapeutic
Relevance |
TP53 |
>50%
all cancers |
Cell cycle
checkpoint/Apoptosis |
Pan-cancer
(highest in ovarian, lung) |
MDM2
inhibitors, p53 reactivators |
KRAS |
~30% all
cancers |
RAS/MAPK
signaling |
Pancreatic
(90%), Colorectal (40%), Lung (25%) |
KRAS G12C inhibitors
(sotorasib, adagrasib) |
PIK3CA |
~20% solid
tumors |
PI3K/AKT/mTOR |
Breast
(45%), Colorectal (15%), Endometrial |
PI3K
inhibitors (alpelisib) |
APC |
80%
colorectal |
Wnt
signaling |
Colorectal,
Gastric |
Wnt
pathway modulators (experimental) |
DNA
Repair Pathway Genes
Gene |
Mutation
Frequency |
Primary
Pathway |
Major
Cancer Types |
Therapeutic
Relevance |
BRCA1/BRCA2 |
5-10%
breast/ovarian |
Homologous
recombination |
Breast,
Ovarian, Prostate, Pancreatic |
PARP
inhibitors (olaparib, niraparib) |
ATM |
5-15%
various |
DNA damage
response |
CLL,
Breast, Prostate |
ATR
inhibitors, PARP inhibitors |
MLH1/MSH2/MSH6/PMS2 |
15%
colorectal |
Mismatch
repair |
Colorectal,
Endometrial, Lynch syndrome |
Immune
checkpoint inhibitors |
Receptor
Tyrosine Kinases & Growth Factors
Gene |
Mutation
Frequency |
Primary
Pathway |
Major
Cancer Types |
Therapeutic
Relevance |
EGFR |
15% lung
adenocarcinoma |
RTK/MAPK
signaling |
Lung,
Glioblastoma, Head/Neck |
TKIs
(erlotinib, osimertinib) |
HER2
(ERBB2) |
20% breast
cancer |
RTK
signaling |
Breast,
Gastric |
Trastuzumab,
T-DM1, TKIs |
ALK |
5% lung
adenocarcinoma |
RTK fusion
proteins |
Lung,
Lymphomas |
ALK
inhibitors (crizotinib, alectinib) |
Chromatin
Remodeling & Epigenetic Regulators
Gene |
Mutation
Frequency |
Primary
Pathway |
Major
Cancer Types |
Therapeutic
Relevance |
ARID1A |
10-50% various |
SWI/SNF
chromatin remodeling |
Ovarian,
Endometrial, Gastric |
Synthetic
lethal approaches |
KMT2D |
20%
lymphomas |
Histone
methylation |
Diffuse
large B-cell lymphoma |
Histone
methyltransferase inhibitors |
IDH1/IDH2 |
70%
gliomas, 20% AML |
Metabolic/Epigenetic |
Glioma,
AML, Cholangiocarcinoma |
IDH
inhibitors (ivosidenib, enasidenib) |
Cell
Cycle Regulation
Gene |
Mutation
Frequency |
Primary
Pathway |
Major
Cancer Types |
Therapeutic
Relevance |
CDKN2A
(p16) |
30-50%
various |
G1/S
checkpoint |
Melanoma,
Pancreatic, Lung |
CDK4/6
inhibitors |
RB1 |
90%
retinoblastoma |
G1/S
checkpoint |
Retinoblastoma,
Small cell lung |
CDK4/6
inhibitors, aurora kinase inhibitors |
CCND1 |
15-20%
breast |
G1/S
progression |
Breast,
Mantle cell lymphoma |
CDK4/6
inhibitors (palbociclib) |
Emerging Drivers
(Recent WGS Studies 2024)
Discovery |
Context |
Primary
Pathway |
Cancer
Types |
Therapeutic
Potential |
74 New
Candidate Genes |
Nature
Genetics 2024 (10,478 genomes) |
RNA
processing, protein degradation |
Pan-cancer
analysis |
Under
investigation |
Non-coding
drivers |
Regulatory
elements, lncRNAs |
Gene
expression regulation |
Multiple
cancer types |
Epigenetic
modulators |
Table 2.
Target (substrate) |
PDB/AlphaFold ID2 |
Keq (M-1)3 |
B (mT)1 |
|
GTPase switch
protein |
|
|
|
|
Hras (GTP) |
PDB 8ELK |
9.3x1010 |
625 |
|
Kras (Raf1 Ras BD) |
PDB 6XHA |
2.8 x 106 |
130 |
|
Protein
tyrosine kinase |
|
|
|
|
c-Abl (ATP) |
AF-P00520-F1 |
8.3 x 104 |
18 |
|
ALK (ATP) |
AF-Q9UM73-F1-v4 |
2.4 x 105 |
28 |
|
BTK (ATP) |
AF-Q06187-F1 |
3.4 x 104 |
43 |
|
EGFR (ATP) |
AF-P00533-F1 |
5.9 x 104 |
28 |
|
HER2 (ATP) |
AF-P04626-F1 |
3.7 x 104 |
23 |
|
c-kit (ATP) |
AF-P10721-F1-v4 |
1.9 x 104 |
18 |
|
SRC (ATP) |
AF-P00523-F1-v4 |
1.2 x 104 |
102 |
|
VEGFR1 (ATP) |
AF-P17948-F1 |
7.7 x 103 |
6 |
|
VEGFR2 (ATP) |
AF-P35968-F1 |
7.7 x 103 |
10 |
|
Protein
serine/threonine kinase |
|
|
|
|
AKT1 (ATP) |
AF-P31749-F1 |
7.6 x 103 |
36 |
|
AKT2 (ATP) |
AF-P31751-F1 |
3.9 x 103 |
53 |
|
ATR (ATP) |
AF-Q13535-F1-v4 |
2.0 x 107 |
28 |
|
Aurora 2
(ATP) |
AF-O14965-F1 |
2.9 x 104 |
30 |
|
Cdk2-PO4/Cyclin A
(ATP) |
PDB 1JST |
4.3 x 104 |
57 |
|
Cdk2/Cyclin E
(ATP) |
PDB 1W98 |
2.8 x 105 |
44 |
|
Cdk4/Cyclin D1 (ATP) |
PDB 2W96 |
2.4 x 103 |
33 |
|
Cdk6/vCyclin
(ATP) |
PDB 1JOW |
1.2 x 105 |
61 |
|
Chk1 (ATP) |
AF-O14757-F1 |
7.1 x 105 |
72 |
|
Chk2 (ATP) |
AF-O96017-F1 |
3.0 x 105 |
30 |
|
pERK1 (ATP) |
PDB 2ZOQ |
3.2 x 105 |
71 |
|
pERK2 (ATP) |
PDB 6OPG |
5.5 x 105 |
63 |
|
GSK3β (ATP) |
AF-49841-F1 |
2.0 x 104 |
44 |
|
MEK1 (ATP) |
AF-Q02750-F1 |
1.8 x 105 |
58 |
|
RAF1 (ATP) |
AF-P04049-F1 |
8.6 x 104 |
36 |
|
Lipid
phosphoinositol kinase |
|
|
|
|
ATM (ATP) |
PDB 7NI6 |
3.4 x 104 |
19 |
|
PIK3CA (ATP) |
AF-P42336-F1 |
5.0 x 105 |
49 |
|
MTOR (ATP) |
PDB 3JBZ |
1.0 x 103 |
15 |
|
Other enzymes |
|
|
|
|
PARP1 (NAD) |
AF-P09874-F1-v4 |
1.3 x 106 |
29 |
|
PARP2 (NAD) |
AF-Q9UGN5-F1-v4 |
5.3 x 105 |
25 |
|
IDH1 (Isocitrate) |
PDB 3INM |
1.5 x 104 |
306 |
|
IDH2
(Isocitrate) |
PDB 5I95 |
1.7 x 105 |
230 |
|
1 Calculation of B (mT) was based on
equations [1]-[6].
2 The dipole moment (μ) of the proteins was calculated from
coordinates provided in the corresponding
PDB or AlphaFold files shown below, and using the Protein Dipole Moments Server . The server is described in Clifford E. Felder, Jaime
Prilusky, Israel Silman, and Joel L. Sussman 2007, " A server and database
for dipole moments of proteins", Nucleic Acids Research, 35,
special Web Servers Issue. https://academic.oup.com/nar/article/35/suppl_2/W512/2922221.
3 The Keq in equation [5] is 1/Kd
for simple protein-ligand interaction, or 1/Km for enzyme substrate
interaction. The catalytic Km is used as a first approximation of
substrate affinity. Kd or Km values were obtained from the following sources:
Hras (GTP): John, J., et al., 1993, Kras (Raf1 Ras BD): Tran, T.H., et
al., 2021, c-Abl (ATP), BTK (ATP), EGFR (ATP), HER2 (ATP), c-kit (ATP), SRC
(ATP), VEGFR1 (ATP), VEGFR2 (ATP), AKT1 (ATP), AKT2 (ATP), Aurora 2 (ATP),
Cdk2-PO4/Cyclin A (ATP), Cdk2/Cyclin E (ATP), Cdk4/Cyclin D1 (ATP), Chk1 (ATP),
Chk2 (ATP), GSK3β (ATP), MEK1
(ATP), RAF1(ATP), ATM
(ATP) & MTOR (ATP): Knight, Z.A. & Shokat, K.M., 2005, ALK (ATP):
Bossi, R.T., et al., 2010, ATR: data from ReactionBiology and Eurofins, Cdk6/vCyclin (ATP): data from ReactionBiology, pERK1 (ATP) & pERK2 (ATP): Petrosino,
M., et al., 2023, PIK3CA (ATP): Maheshwari, S., et al. 2017,
PARP1 (NAD) & PARP2 (NAD): Thorsell A-G., et al., 2016, IDH1 (Isocitrate):
Uniprot O75874, IDH2 (Isocitrate): Uniprot P48735
Acknowledgement: The author thanks Bill Windsor for
providing some of the literature cited in this review.
Dedication:
To my brother Kiet,
who in heaven will know why.
Comments
Post a Comment