Cancer incidence is projected to rise dramatically, potentially adding up to 32 million new cases annually by 2050.1 This surge is fueling remarkable innovation in oncology, with more than 100 new treatments anticipated to enter the market within the next five years.2 As a dominant sector within the global pharmaceutical market, oncology generated $223 billion in sales in 2023, and is forecasted to double by 2028.2 As we celebrate the wave of new therapies today, it's essential to reflect on the remarkable progress made over the past century.
1950s – Chemotherapy: The First Frontier
Chemotherapy represents one of the earliest oncology modalities. In 1946, researchers discovered that nitrogen mustards — originally developed for chemical warfare — could effectively kill tumor cells.3 Nitrogen mustard, eventually known as mechlorethamine, is an alkylating agent capable of damaging cancer cell DNA, preventing replication and leading to cell death. Approved in 1949, this discovery laid the groundwork for modern chemotherapies.
1960–1980s – Genetic Discoveries Driving Innovation
The subsequent decades brought groundbreaking discoveries, largely driven by advances in genetics. In 1960, scientists David Hungerford and Peter Nowell identified the "Philadelphia chromosome," a translocation shift between chromosomes that created the BCR-ABL fusion gene, associated with higher cancer risks.3 Later in 1979, the critical tumor suppressor gene p53 — today known as the most frequently mutated gene in cancer — was discovered.4
Further advances followed, such as the identification of the 'neu' oncogene in rats in 1984, eventually leading to the discovery of HER2, a gene implicated in approximately 25% of aggressive breast cancers.4 Shortly thereafter, researchers isolated and sequenced BRCA2, a gene strongly linked to breast and ovarian cancers, opening new avenues for targeted treatment.
1990–2000s – The Era of Targeted Therapies
These genetic discoveries paved the way for targeted therapies, significantly improving upon traditional chemotherapy by precisely targeting cancer-specific pathways while sparing healthy cells. In 2001, the FDA approved imatinib (GleevecⓇ), the first targeted therapy designed to inhibit the BCR-ABL protein, revolutionizing the treatment of chronic myeloid leukemia. Once a fatal diagnosis, chronic myeloid leukemia is now a manageable condition.3
Concurrently, monoclonal antibodies emerged as a powerful therapeutic modality. Rituximab, approved in 1997 for non-Hodgkin’s lymphoma, was the first monoclonal antibody therapy. In 1998, trastuzumab (HerceptinⓇ), targeting HER2+ breast cancer, was approved and significantly reduced recurrence rates by up to 60%, dramatically reshaping breast cancer management.4
2010–2016 – The Rise of Immunotherapies
This period saw immunotherapy rise to prominence, spearheaded by immune checkpoint inhibitors. James Allison's pioneering work demonstrated that blocking the immune checkpoint CTLA-4 could activate immune responses against tumors, leading to the approval of ipilimumab (YervoyⓇ) in 2011 for advanced melanoma. This breakthrough inspired additional checkpoint inhibitors targeting different proteins. This also led to the development of combination therapies like ipilimumab and nivolumab (OpdivoⓇ), now standard treatment protocols for advanced melanoma.1
Simultaneously, cancer vaccines emerged prominently, exemplified by Gardasil, approved in 2006 for human papillomavirus (HPV)-induced cervical cancer. Providing nearly 90% protection against cervical cancer, Gardasil has dramatically reduced the incidence of one of the historically deadliest cancers affecting women. Now, Gardasil is offered for free in schools in 140 countries globally.5
Additionally, this era introduced oncolytic virus therapies, notably talimogene laherparepvec (T-VEC), approved in 2015 for melanoma.5 These therapies harness genetically modified viruses to selectively infect and lyse tumor cells, activating immune responses against cancers, signaling oncology's transition into the age of genetic manipulation and precision medicine.
By 2016, immunotherapy firmly established itself as the fourth pillar of cancer treatment, alongside surgery, radiation, and chemotherapy, earning recognition as the 'Clinical Advance of the Year' by the American Society of Clinical Oncology.6
2017–Present – The Beginning of Cell and Gene Therapies
Recent years marked the development of revolutionary cell and gene therapies. Recognizing the inherent limitations of natural immune cells in targeting cancers, scientists engineered chimeric antigen receptor (CAR)-T cells — immune cells genetically modified to precisely recognize and destroy cancer cells. The FDA approvals of tisagenlecleucel (KymriahⓇ) and axicabtagene ciloleucel (YescartaⓇ) in 2017 for blood cancers exemplified this innovation, creating what many termed "living drugs."6
2020 and Beyond: Moving Toward Precision Oncology
Over 70 years have passed since the advent of chemotherapy, and oncology treatments have undergone dramatic transformation in this period. Modern cancer therapies have evolved from broadly cytotoxic approaches toward increasingly precise targeting of specific molecular pathways. Therapeutic strategies have progressed from indiscriminate cancer-cell elimination to harnessing the body's own immune response via monoclonal antibodies like rituximab. Moreover, monotherapies are being supplemented by combinational approaches designed for improved efficacy. The drug types themselves have grown progressively complex, transitioning from traditional small-molecule inhibitors toward biologically complex large molecules. Previously "undruggable" molecular targets are now within reach through gene-editing strategies, cell-based therapies, and therapeutic cancer vaccines.1
Emerging Modalities on the Horizon
Although traditional therapeutic approaches still constitute the majority of the oncology market, novel therapeutic modalities such as antibody–drug conjugates (ADCs), bispecific antibodies, and radioligand therapies are rapidly gaining prominence. Clinical trial activity for these innovative modalities has increased markedly, with a notable 27% growth since early 2023 alone.2 Market forecasts indicate a continued rise, predicting that by 2028 these modalities could collectively generate approximately $37 billion in annual global sales.2
Antibody–Drug Conjugates
ADCs have rapidly become a central modality in oncology, recently commanding nearly 80% of oncology-related pharmaceutical deal values, according to IQVIA Pharma Deals (2023).2 Structurally, ADCs are composed of a monoclonal antibody chemically linked to a potent cytotoxic drug payload. Often described as “biological missiles,” ADCs selectively target cancer cells, delivering highly potent agents directly to tumor sites, thereby maximizing efficacy and limiting off-target toxicity.7
Since the FDA approval of the first ADC — gemtuzumab ozogamicin (MylotargⓇ) — in 2000, the development of ADCs has accelerated remarkably, now comprising over 850 ongoing clinical trials and 15 approved drugs globally.2 Solid tumors constitute the primary focus of ADC research, with prominent antigen targets including HER2, TROP2, and B7-H3.7 Ongoing research efforts aim to expand the therapeutic potential of ADCs by identifying novel tumor-specific antigens and refining linker–payload chemistries to optimize safety and clinical effectiveness.
Radioligand Therapies
Radioligand therapies (RLTs) represent a novel and rapidly growing therapeutic approach that combines the precision targeting of ligands or antibodies with the potency of radioactive isotopes. Unlike traditional radiotherapy, which often risks collateral damage to surrounding healthy tissues, radioligand therapies precisely deliver targeted radiation to cancer cells expressing specific receptors or biomarkers. This molecularly targeted radiation allows for maximal therapeutic impact with minimal off-target side effects, representing a significant advancement in oncology precision medicine.
The FDA's recent approvals, including lutetium Lu 177 vipivotide tetraxetan (PluvictoⓇ) in 2022 for prostate cancer, underscore the clinical promise of radioligand therapies.2 Future innovations in this field will likely involve the discovery of novel molecular targets, development of safer and more effective radioisotopes, and refinement of ligand-receptor specificity, further expanding the scope and effectiveness of this exciting therapeutic class.1
Bispecific Antibodies
Bispecific antibodies represent an innovative class of therapeutics engineered to simultaneously bind two distinct molecular targets, typically involving tumor cells and immune cells. By bridging immune effectors — often cytotoxic T cells — with cancer cells, bispecific antibodies facilitate targeted immune-mediated killing, enhancing antitumor efficacy while reducing systemic toxicity. The capability of bispecific antibodies to engage immune cells directly to tumor cells offers a compelling therapeutic advantage, distinct from traditional monoclonal antibodies.
Clinical development of bispecific antibodies has accelerated dramatically, with notable FDA approvals such as blinatumomab (BlincytoⓇ), the first bispecific T cell engager (BiTE), approved in 2014 for acute lymphoblastic leukemia.5 Since then, bispecific antibodies have expanded rapidly in clinical trials across a wide spectrum of hematologic malignancies and solid tumors. Promising therapeutic targets include CD3-based T cell engagers as well as dual blockade of tumor-associated antigens like CD20, BCMA, and HER2. Looking forward, continued innovation in bispecific antibody engineering, including advancements in antibody stability, dosing optimization, and the identification of novel dual-target combinations, is poised to significantly shape the future oncology landscape.2
Conclusion
The evolution of oncology treatments over the past century vividly illustrates how innovation drives transformative progress in cancer care. From the early days of chemotherapy through immunotherapies and today's precision-guided modalities, each advancement has reshaped our approach to fighting cancer. The future of oncology holds enormous promise, offering renewed hope for millions of patients and underscoring the extraordinary potential of scientific discovery and clinical innovation to conquer one of the deadliest human diseases.
Our parent company, That’s Nice, is committed to supporting the companies and innovators driving the next wave of pharma and biotech innovation. To celebrate That’s Nice’s 30th anniversary, Pharma’s Almanac is diving into 30 groundbreaking advancements, trends, and breakthroughs that have shaped the life sciences, highlighting the industry-defining milestones our agency has had the pleasure of growing alongside. Here’s to 30 years of innovation and the future ahead!
References
1. Eskandar, Kirolas. “Emerging trends in oncology: a comprehensive literature review.” Surgery and Oncology. 14: 59–68 (2024).
2. Gores, Markus and Stefan Lutzmayer. “Breaking New Ground: Advancing Cancer Care with Novel Therapeutic Modalities.” IQVIA. 2024.
3. “The History of Cancer Therapy, an Interactive Timeline.” Scientific American. Accessed 8 Apr. 2025.
4. “Milestones in Cancer Research and Discovery.” National Cancer Institute. 21 Feb. 2025.
5. Liu, Beilei, et al. “Exploring treatment options in cancer: Tumor treatment strategies." Signal Transduction and Targeted Therapy. 9: 1–44 (2024).
6. Brodsky, Arthur D. “The Decade of Immunotherapy: Highlights from the 2010s.” Cancer Research Institute. 30 Dec. 2019.
7. Fu, Zhiwen, et al. “Antibody drug conjugate: the ‘biological missile’ for targeted cancer therapy.” Signal Transduction and Targeted Therapy. 7: 93 (2022).