Cancer treatment has changed dramatically over the past two decades. One of the biggest breakthroughs has been the development of medicines that focus on a single, well-defined target inside a cancer cell. These medicines, called single-target compounds, are designed to switch off the specific molecule or pathway that helps a tumor survive and grow.
For many patients, this marks a shift from traditional chemotherapy, which works broadly across the body, to precision therapy that is tailored to the biology of their own cancer.
These treatments are not just scientific innovations. They represent hope for patients with cancers that were once considered untreatable, and they demonstrate what is possible when medicine becomes more personalized.
How Single-Target Cancer Medicines Are Discovered
The development of these drugs begins long before a patient receives them. Researchers first identify a molecule inside cancer cells that plays a key role in driving the disease. This molecule could be a mutated protein, a surface receptor, or a signaling enzyme that helps cancer cells divide uncontrollably[1].
Scientists then test thousands of compounds through laboratory screening and computer-based simulations to find one that can block this target safely and effectively. This stage has become far more advanced with the rise of genomics. Today, next-generation sequencing can reveal the exact mutations in a patient’s tumor, allowing researchers to focus on targets that matter most[2].
One of the best known examples is imatinib, developed for chronic myeloid leukemia. This drug was designed to silence a specific abnormal protein called BCR-ABL. What began as a weak lead molecule was optimized step by step until it became a therapy that transformed leukemia survival rates worldwide[3].
This story is not unique. Similar advances have been observed across lung cancer, breast cancer, melanoma, and many other malignancies, proving that when you find the right target, you can reshape the entire course of a disease.
How These Medicines Work Inside the Body
Single-target compounds fall into two main categories: small molecule inhibitors and monoclonal antibodies. Both work differently but share the same mission: to shut down the pathways tumors rely on.
Small molecule inhibitors
These medicines are tiny enough to enter cancer cells and block internal proteins known as kinases. Kinases act like switches inside the cell, turning signals on and off. When mutated, they can get stuck in the “on” position, driving cancer cells to multiply.
By blocking the kinase at its active site or a nearby allosteric site, small molecule inhibitors prevent the signals that drive cancer growth. This slows or stops the disease at the molecular level.
Monoclonal antibodies
These treatments target receptors or proteins on the surface of cancer cells. They can stop growth signals from being sent, mark cancer cells for destruction by the immune system, or trigger cell death directly.
Rituximab, trastuzumab, and many others have proven how powerful targeted antibodies can be when a tumor expresses the right marker[1].
When Single-Target Compounds Work Best
These medicines have had the greatest impact in cancers where a specific mutation or protein plays a central role. Key examples include:
• Chronic myeloid leukemia: Imatinib revolutionised survival, helping more than 80 percent of patients live for many years with good quality of life[3].
• Lung cancer: Patients with EGFR mutations or ALK fusions often respond dramatically to drugs specifically designed for these abnormalities, with response rates exceeding 60 percent[4].
• HER2-positive breast cancer: Trastuzumab improves survival when added to chemotherapy and reduces the risk of recurrence over the long term[5].
• Melanoma: Blocking BRAF and MEK simultaneously has extended survival for patients with BRAF-mutant disease[6].
These are examples of personalised medicine at its best, where knowing a single mutation can reshape treatment decisions entirely.
Why Resistance Still Happens
As effective as targeted drugs can be, they are not perfect. Many patients eventually develop resistance. Cancer cells are highly adaptive and can find new ways to survive even after their main driver is blocked.
Resistance can arise because the target mutates, the cancer activates a backup pathway, or the tumor evolves into a different cell type altogether. In some cases, small pockets of resistant cells already exist before treatment even begins.
For patients, this can feel discouraging, especially after an initially strong response. But understanding resistance has helped guide the next phase of progress.
Combination Strategies: The Future of Single-Target Therapy
To overcome resistance, researchers are now combining targeted drugs with others, with immunotherapy, or with novel agents such as SHP2 inhibitors. These combinations aim to shut down multiple pathways at once, leaving the cancer with fewer escape routes.
This approach has already shown success. BRAF and MEK inhibitors used together in melanoma deliver deeper and longer responses. In colorectal cancer with BRAF mutations, combinations involving EGFR and BRAF inhibitors outperform single-agent therapy[7].
Even more advanced strategies are being explored through artificial intelligence, high-throughput screening, and real-time genomic monitoring.
For patients, this means a future defined by treatments that are increasingly personalized, increasingly effective, and increasingly designed around the uniqueness of their own cancer.
At Helix BioPharma, we are committed to advancing this vision.
Our research focuses on therapies that directly address the molecular vulnerabilities of difficult cancers, while also overcoming the barriers that limit current targeted treatments. By integrating scientific insight with patient-centred innovation, our goal is to help move precision oncology from possibility to reality, and to bring better options to those who need them most.
Ref:
1. Min HY, Lee HY. Molecular targeted therapy for anticancer treatment. Exp Mol Med. 2022 Oct;54(10):1670-1694. doi: 10.1038/s12276-022-00864-3. Epub 2022 Oct 12. PMID: 36224343; PMCID: PMC9636149.
2. Doostmohammadi A, Jooya H, Ghorbanian K, Gohari S, Dadashpour M. Potentials and future perspectives of multi-target drugs in cancer treatment: the next generation anti-cancer agents. Cell Commun Signal. 2024 Apr 15;22(1):228. doi: 10.1186/s12964-024-01607-9. PMID: 38622735; PMCID: PMC11020265.
3. Baran Y, Saydam G. Cumulative clinical experience from a decade of use: imatinib as first-line treatment of chronic myeloid leukemia. J Blood Med. 2012;3:139-50. doi: 10.2147/JBM.S29132. Epub 2012 Nov 16. PMID: 23180974; PMCID: PMC3503471.
4. Leonetti A, Sharma S, Minari R, Perego P, Giovannetti E, Tiseo M. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br J Cancer. 2019 Oct;121(9):725-737. doi: 10.1038/s41416-019-0573-8. Epub 2019 Sep 30. PMID: 31564718; PMCID: PMC6889286.
5. Early Breast Cancer Trialists’ Collaborative group (EBCTCG). Trastuzumab for early-stage, HER2-positive breast cancer: a meta-analysis of 13 864 women in seven randomised trials. Lancet Oncol. 2021 Aug;22(8):1139-1150. doi: 10.1016/S1470-2045(21)00288-6. PMID: 34339645; PMCID: PMC8324484.
6. Priantti JN, Vilbert M, Madeira T, Moraes FCA, Hein ECK, Saeed A, Cavalcante L. Efficacy and Safety of Rechallenge with BRAF/MEK Inhibitors in Advanced Melanoma Patients: A Systematic Review and Meta-Analysis. Cancers (Basel). 2023 Jul 25;15(15):3754. doi: 10.3390/cancers15153754. PMID: 37568570; PMCID: PMC10417341.
7. Liu M, Yang X, Liu J, Zhao B, Cai W, Li Y, Hu D. Efficacy and safety of BRAF inhibition alone versus combined BRAF and MEK inhibition in melanoma: a meta-analysis of randomized controlled trials. Oncotarget. 2017 May 9;8(19):32258-32269. doi: 10.18632/oncotarget.15632. PMID: 28416755; PMCID: PMC5458282.