19707 items found
ASCO 2024 - Chris Haqq interview with Jonah Comstock
Oncology

ASCO 2024 - Chris Haqq

ASCO isn’t just a show for top 10 pharmas and Phase 3 readouts; it’s …

Oncology trials: Top 5 trends to watch

Advanced Clinical logo

Oncology trials: Top 5 trends to watch

From innovative trial designs to groundbreaking imaging techniques, oncology studies are rapidly evolving. Advanced Clinical’s VP of oncology, Ravi Karra MD, MBA, explores the top trends shaping the future of oncology clinical trials.

The oncology clinical trial landscape is undergoing a profound transformation, driven by the rapid pace of pharmaceutical and biopharmaceutical innovation in cancer research. As new and advanced therapy options continue to emerge, such as cell and gene therapies and immunotherapies, the enthusiasm surrounding oncology clinical trials and the promise they hold for patients is reaching new heights. 

The emergence of innovative new drug classes, along with improved imaging techniques, is reshaping the way clinical trials are designed, conducted, and analysed. This ultimately contributes to steady improvements in overall survival (OS) and progression-free survival (PFS) for patients across a wide range of cancer types.

Here, we delve into the key trends shaping oncology clinical trials and examine the innovative trial designs and cutting-edge techniques that are set to optimise patient outcomes and accelerate the path to new cancer treatments.

 

Trend 1: Trial designs are changing


The traditional linear and sequential approaches to clinical trials are giving way to innovative, adaptive, and flexible methodologies. These new trial designs are crucial in addressing the complex demands of modern oncology treatments, aiming to accelerate the development of life-saving therapies. Adaptive trial designs allow researchers to dynamically modify trial parameters based on interim results, utilising real-time data. This flexibility minimises the number of required participants and shortens trial durations, making the process more efficient and effective. If early efficacy data shows a higher-than-anticipated response rate, researchers can adjust the sample size accordingly, reducing unnecessary exposure to ineffective treatments and expediting the development timeline.

Basket trials evaluate a drug's effectiveness against a specific mutation across multiple cancer types. This approach enhances the applicability of findings, enabling a single treatment to be explored in various contexts. For example, a drug targeting the BRAF mutation can be concurrently tested in patients with melanoma, colorectal cancer, and thyroid cancer – consolidating research efforts and potentially accelerating the availability of effective treatments. 

Just as basket trials explore a drug's potential across different cancers, umbrella trials take a complementary approach. Umbrella trials involve testing multiple drugs on various mutations within a single cancer type. This method streamlines the process for complex cases by combining multiple treatment pathways under one protocol. In colorectal cancer, for instance, diverse treatments targeting various genetic mutations can be evaluated simultaneously, fostering a comprehensive understanding of the disease and its potential therapies.

Trend 2: Transforming early-stage clinical development


Complementing the paradigm shift in clinical trial designs, early-stage clinical development is evolving to incorporate more sophisticated and targeted approaches, focusing on rapidly identifying promising drugs based on patient-specific factors and biological markers. 

Traditionally, Phase 1 oncology trials solely evaluated maximum tolerated dosages of harsh chemotherapies. But that model is evolving for today's targeted therapies and immunotherapies. What used to be Phase 1 – purely a safety trial assessing maximum doses – has moved on. The new focus is on determining the optimal biological dose: the level that can effectively attack a patient's tumour while remaining tolerable.

Rather than just toxicity, these new biomarker-driven early trials aim to identify the ideal therapeutic dose matched to each patient's specific biology from the start. Novel designs like the Bayesian Optimal Interval approach are replacing the former "3+3" standard, allowing smarter and faster dose evaluations, aligned with precision medicine.

The shift in early trial methodologies reflects the broader transformation occurring in oncology drug development. Biologics have gained significant traction, encompassing innovative cell and gene therapies where genes are manipulated ex vivo or in vivo. Treatments like CAR-T cell therapies targeting a patient's own re-engineered immune cells and drugs inhibiting specific mutations are now routinely explored from the earliest clinical stages.

By evolving Phase 1 methodologies to focus on optimal biological dosing and patient biomarkers upfront, we can more efficiently prioritise modern oncology's most promising therapeutic candidates. More innovative early trial designs match the increased scientific complexity, aligning these crucial initial human studies with a personalised future for cancer therapy. Reforming our early development paradigms will be essential to translating advances in biology into patient impacts.

Trend 3: The rise of new drug classes


While chemotherapies were once our primary arsenal, oncology has radically expanded its therapeutic horizons over the past couple of decades. We have transitioned from simply relying on small molecule chemicals to harnessing the power of large, complex biological molecules and even re-engineering living cells themselves.

What once seemed like science fiction is increasingly becoming standard-of-care at major cancer centres across Western Europe and North America, particularly for haematological malignancies. Novel therapeutic classes, such as immunotherapies, are leading this transformation. 

This shift is reflected in regulatory designations, such as the European Medicines Agency's "Advanced Therapy Medicinal Products" (ATMPs) category, which encompasses groundbreaking modalities, including cell and gene therapies involving ex vivo or in vivo manipulation of patient genes or cells. Under this approach, a patient's immune cells can be extracted, bioengineered to selectively recognise and attack their specific cancer, and then reinfused back into the body as a living therapeutic to eradicate tumours from within.

Notably, immune checkpoint inhibitors function as a sort of "brake release" - allowing the body's own T-cells and other defences to vigorously attack cancer after the tumour's immune-evasion mechanisms are blocked. Since malignancies originate from the patient's own cells, they employ sophisticated camouflage tactics. Checkpoint inhibitors strip away that disguise so that the cancer can no longer hide.

Alongside immunotherapies, we have seen a boom in precision-targeted therapies designed to hit specific genetic drivers, like BRCA mutations, in breast cancer. When a patient's tumour harbours that molecular aberration, these rational drugs can provide exquisitely targeted and effective treatment by directly inhibiting the root cause.

The oncology pipeline has rapidly expanded beyond our former chemotherapy horizons, from re-engineered living cells serving as therapeutics to genomically targeted magic bullets and immunomodulators. Continued research, development, and innovation across these diverse modalities will be vital for sustaining transformative advances against cancer.

Trend 4: Innovation in imaging techniques


While techniques like X-rays, ultrasounds, CT scans, and MRIs remain vital for detecting tumour masses and monitoring changes over time, these anatomical imaging modalities provide limited insight into the underlying tumour biology and response to treatment. The focus has been on visualising physical characteristics like mass dimensions and location.

This landscape is evolving with the emergence of molecular and functional imaging techniques that can visualise tumours at the molecular and metabolic levels. For example, positron emission tomography (PET) scans using radiolabelled glucose tracers like 18F-FDG can highlight the distribution of viable, metabolically active malignant cells based on their elevated glucose uptake compared to surrounding tissue.

Molecular imaging also enables more precise cancer phenotyping and targeted treatment selection. For prostate cancer specifically, PET tracers targeting prostate-specific membrane antigen (PSMA) overexpressed on malignant cells enable visualisation of disease based on this biomarker phenotype. Rather than simple shape estimates, these scans map the precise distribution and burden of viable PSMA-expressing lesions throughout the body with high resolution.

Beyond visualising metabolic activity, molecular imaging techniques can expose other critical cancer hallmarks, such as angiogenesis, hypoxia, proliferation, and treatment resistance mechanisms. Information that was previously inaccessible is now discernible through these advanced, mechanism-based visualisation approaches.

As molecular profiling and biomarker-guided therapies become increasingly integral to personalised cancer care, these advanced imaging platforms complement anatomical techniques by providing additional context about each patient's specific tumour biology and treatment response patterns. Integrating molecular and anatomical visualisation methods enhances diagnostic, staging, and treatment monitoring capabilities across oncology.

Trend 5: Optimising existing cancer treatments to suit patient profiles


Radiotherapy Treatments for cancer

Historically, the relatively small patient populations impacted by rare cancer types posed challenges for driving substantial research and drug development investments from biopharmaceutical companies. With high costs and extended timelines for oncology product development, companies generally needed to project broad potential markets to ensure financial viability following approval.

However, that landscape is shifting, due to new regulatory policies specifically aimed at incentivising therapies for rare diseases. Agencies like the FDA and EMA have implemented programmes granting extended market exclusivity periods of eight to ten years to sponsors developing rare disease treatments. These protections provide sufficient time to establish new medicines and recoup investments before biosimilar competition emerges.

As a result, clinical trials are increasingly open to explore both novel agents and existing approved drugs that may have utility for new rare cancer indications. Strategies like repurposing or repositioning can help rapidly expand treatment options for underserved populations by circumventing portions of the standard drug development pathway.

Optimising available treatments by choosing the best sequence of first, second, and further lines of therapy, alone or in combination, based on patient-specific factors, represents a truly personalised approach. Ongoing efforts to refine and enhance treatments for widespread cancers, like colorectal and breast cancer, are critical and illustrated by new combination treatments and better sequencing of treatment lines, which offer improved outcomes with fewer side effects. For example, integrating novel targeted therapies with traditional chemotherapy regimens can enhance efficacy while minimising toxicity.

Driven by these new financial incentives and scientific approaches, the oncology pipeline is expanding to encompass more therapies for rare tumour types that were historically deprioritised. This policy-driven shift is catalysing a treatment landscape transformation for patients with uncommon cancers.

About Advanced Clinical

Advanced Clinical is a clinical development and strategic resourcing organisation committed to providing a better clinical experience across the drug development journey. Our goal is to improve the lives of all those touched by clinical research – approaching each opportunity with foresight, character, resilience, and innovation. Based on decades of experience, we help our clients achieve better outcomes by conducting candid conversations and anticipating potential issues through our customised solutions. 

Visit our website to learn more: www.advancedclinical.com.

About the author

Dr Ravi Karra is a clinical developer in oncology with over 20 years’ experience in senior medical roles in academia and industry including pharma and CROs. He is an expert in brain tumours, melanoma, head and neck cancer, and has been a part of several transformative trials like adjuvant Ipilimumab therapy in Stage III melanoma (Eggermont AMM et al. Lancet Oncol. 2015 May;16(5):522-30) and comparing TPF to PF in patients with unresectable squamous cell carcinoma of the head and neck (Vermorken J. B et al. J. Clin. Oncol.2011 29:15_suppl, 5530-5530). He also led trials in rare and difficult to treat cancers like metastatic uveal melanoma (Leyvraz S et al. Ann Oncol. 2014 Mar; 25(3): 742–746). 

He has actively participated in designing and developing innovative trial methods using translational research including molecular and imaging biomarkers, as well as the "Window of Opportunity" concept across tumour types, including rare tumours such as glioblastoma and salivary glands. Dr Karra has extensive experience dealing with the FDA, EMA, and payers for reimbursement, and has been part of successful NDA and ANDA applications and market launches.

Sign up

Supercharge your pharma insights: Sign up to pharmaphorum's newsletter for daily updates, weekly roundups, and in-depth analysis across all industry sectors.

Click on either of the images below for more articles from this edition of Deep Dive: Oncology 2024

Transforming oncology: The rise of targeted radiopharmaceuticals

Putnam logo

Transforming oncology: The rise of targeted radiopharmaceuticals

The advent of targeted radiopharmaceuticals is revolutionising the field of oncology. These innovative therapies integrate systemically delivered radiotherapy into diagnostic and personalised medicine strategies, offering a new frontier in cancer treatment. Unlike traditional therapies, targeted radiopharmaceuticals deliver lethal doses of radiation directly to malignant cells, minimising damage to healthy tissues. This precise targeting is achieved through radioisotope payloads linked to molecules or antibodies that hone in on cancer cells.

Significant progress in this field is evident with the FDA and EMA approval of two groundbreaking treatments: Lutathera for gastroenteropancreatic neuroendocrine tumours (GEP-NETs) and Pluvicto for metastatic castration-resistant prostate cancer (mCRPC). These therapies, now part of Novartis’s portfolio, generated $1.8 billion in revenue in 2023, with Pluvicto alone projected to exceed $3 billion globally. This financial success underscores radioligand therapeutics' immense commercial and therapeutic potential, signalling their emergence as a pillar of modern oncology.

The growing pipeline and strategic partnerships

The expanding pipeline of targeted radiopharmaceuticals has sparked a flurry of business development and licensing activities among major pharmaceutical companies. Over 30 innovators have advanced over 40 assets into clinical development, attracting significant interest from leading global pharmaceutical players. These companies are eager to enhance their portfolios with these innovative agents or to strengthen their in-house radiopharmaceutical capabilities through high-potential assets. Over 20 significant partnering deals have been reported in the past two years alone.

Notable partnerships and acquisitions include:

  • Novartis: In addition to Lutathera and Pluvicto, Novartis has licensed an FAP targeting construct from 3B Pharmaceuticals and partnered with Bicycle Therapeutics to develop protein ligands for oncology.
  • Bayer: Bayer has expanded its radiopharmaceutical portfolio by acquiring Noria and PSMA Therapeutics, gaining access to PSMA (prostate) targeting agents. They have also entered a strategic collaboration with Bicycle Therapeutics to develop additional agents using proprietary, synthetic peptide carriers.
  • Eli Lilly: The acquisition of Point BioPharma has provided Eli Lilly access to pivotal programmes targeting PSMA (mCRPC) and SSTR (GEP-NETs), as well as an early-phase programme targeting FAPα (PNT2004).
  • Bristol Myers Squibb (BMS): BMS's acquisition of RayzeBio for approximately $4.1 billion includes the late-stage asset RYZ101, an actinium-based RLT for GEP-NETs, and RayzeBio's manufacturing facility.
  • Telix Pharmaceuticals: Telix has proposed acquiring QSAM Biosciences and its lead asset, CycloSam (Samarium-153-DOTMP), a radiopharmaceutical for bone cancer or metastasis.

Key development and innovation trends

Several key trends are driving the adoption and clinical value of radiopharmaceuticals in oncology. These trends include expanding indications to address unmet needs in hard-to-treat tumours, improving adverse event profiles, enhancing efficacy, and integrating into combination therapies.

Expansion to hard-to-treat tumours

With proof-of-concept established in GEP-NETs and mCRPC, radiopharmaceutical development has expanded to target over 15 malignancies across solid and haematologic tumours. Innovations are focusing on novel targets not yet fully explored by other treatment modalities, such as hexokinase 2 (HK2), neurotensin receptor 1 (NTSR1), and gastrin-releasing peptide receptor 1 (GRPR1).

Improving adverse event profiles

Next-generation targeted radiotherapeutics aim to enhance radioisotope targeting to improve efficacy and reduce toxicity. Innovations include:

  • Radio DARPin constructs: These genetically engineered antibody mimetic proteins offer high specificity and affinity for target proteins, potentially reducing uptake by healthy tissues and organs, such as the kidneys.
  • Tumour microenvironment (TME) exploitation: Constructs that exploit unique features of the TME, such as lower pH, greater hypoxia, or specific enzymatic activity, to activate or enhance ligand/small molecule or monoclonal antibody (mAb)-mediated binding.
  • Single domain antibodies and nanobodies: These exhibit shorter blood half-lives, enhanced tumour uptake, and superior binding affinity and specificity for target biomarkers.

Enhancing efficacy

Harnessing the greater lethality of alpha-emitting radioisotope payloads represents a significant advancement in radiopharmaceuticals. Alpha emitters deliver high-energy decay with limited tissue penetration, providing potent radiation while minimising exposure to healthy tissues. The emergence of alpha-emitting radioisotopes, such as Actinium-225, Astatine-211, and Lead-212, may reduce reliance on beta emitters, though supply issues persist. Additionally, alternative beta emitters like Copper-67, Iodine-131, and Terbium-161 can diversify radioisotope sources and mitigate supply-side pressures.

black pencils with strong vibrant colours

Combination therapies

The potential for synergistic effects drives the transition from monotherapy to combination therapy. Rationally designed combination therapies include:

  • DNA repair inhibitors: Such as PARP inhibitors, which enhance the efficacy of radiotherapy.
  • Immune checkpoint inhibitors: Anti-PD-(L)1 agents that leverage radiotherapy-induced neo-antigens to boost immune response.
  • Chemotherapeutics: Established radiosensitisers in chemoradiation protocols that enhance the effects of radiotherapy.
Network

Addressing radiopharmaceutical-specific challenges

Developing a differentiated radiotherapeutic portfolio is crucial, but addressing specific challenges related to radiopharmaceuticals is equally important. These challenges include:

Supply chain

Establishing a robust supply chain for medical-grade isotopes is critical. The radio-decay characteristics of these substrates and the anticipated increase in demand underscore the importance of reliable sourcing.

Manufacturing and distribution

Optimising manufacturing and distribution strategies is essential. These strategies might involve centralised, localised (third-party radiopharmacies), or mixed approaches to address geographical and territorial challenges.

Prescriber network

Expanding and realigning field medical and sales teams to engage, educate, and support multidisciplinary care teams, including radiation oncologists and treatment centres capable of administering radiopharmaceuticals, is necessary.

 

Network

In summary

The rapid advancement of targeted radiopharmaceuticals signifies a transformative shift in oncology, offering new hope for patients with difficult-to-treat cancers. By delivering precise radiation directly to malignant cells while sparing healthy tissue, these therapies are poised to revolutionise cancer treatment. The success of pioneering treatments like Lutathera and Pluvicto, coupled with the burgeoning pipeline of over 40 clinical-stage assets, underscores the vast commercial and therapeutic potential of radioligand therapies.

Strategic acquisitions and partnerships among major pharmaceutical companies, such as those by Novartis, Bayer, Eli Lilly, and Bristol Myers Squibb, highlight the intense interest and investment in this field. These collaborations are not only expanding the range of available treatments but also driving innovation in targeting mechanisms and combination therapies. By addressing challenges unique to radiopharmaceuticals, including supply chain logistics, manufacturing, distribution, and prescriber education, the industry is paving the way for more widespread adoption and integration into standard oncology care.

Looking forward, the integration of next-generation targeted radiotherapeutics into combination regimens promises to enhance efficacy and expand their use across various stages of cancer treatment. Innovations such as alpha-emitting radioisotopes and novel targeting constructs are expected to further improve clinical outcomes, reduce adverse events, and provide new avenues for treating a broader range of malignancies.

As the clinical evidence continues to validate the efficacy and safety of these therapies, targeted radiopharmaceuticals are poised to become a cornerstone of modern oncology. The ongoing development and strategic investments in this field reflect a commitment to improving patient outcomes and addressing unmet needs in cancer care. The future of oncology is bright, with targeted radiopharmaceuticals leading the charge toward more personalised, more effective, and less invasive cancer treatments.

The emerging era of targeted radiopharmaceuticals represents a paradigm shift in the treatment of cancer. Through continued innovation, strategic partnerships, and a focus on addressing key challenges, these therapies have the potential to become a central pillar in the fight against cancer. 

 

About Putnam

Putnam logo

Putnam, an Inizio Advisory company, is a leading scientific and strategic consulting partner that helps the global life science community make confident decisions, build value, and bring life-changing innovations to clinicians and patients. For more than 30 years, our rigorous, bespoke approach and globally diverse team have delivered unrivalled depth across therapeutic areas, business functions, geographic markets, healthcare sectors, and technology platforms to maximise the human impact and commercial success of client innovations.

www.putassoc.com

Vikram Gosainv

About the author

Vikram Gosain is a partner in Putnam’s London office and is focused on expanding Putnam’s commercial strategy practice in the UK and Europe.

He brings over 20 years of consulting experience having worked with multiple clients globally to make more informed decisions related to asset, disease, and portfolio strategy. Gosain’s core areas of focus include commercial opportunity assessments, early pipeline strategy, BD&L (Business Development & Licensing) due diligence and portfolio strategy. He has worked across several therapeutic areas specialising in oncology, immunology, rare diseases, cardiovascular, metabolic, and renal diseases. His previous roles include corporate development strategy and banking.

Gosain earned his Bachelor’s and MBA from the Lubin School of Business at Pace University and was inducted to the Beta Gamma Sigma honour society.

Research contributions from Andris Ortmanis, senior director, and Anita Zutshi, associate director.

Sign up

Supercharge your pharma insights: Sign up to pharmaphorum's newsletter for daily updates, weekly roundups, and in-depth analysis across all industry sectors.

Click on either of the images below for more articles from this edition of Deep Dive: Oncology 2024

A history of: Mapping breast cancer’s journey from past to present

A history of: Mapping breast cancer’s journey from past to present 

Breast cancer has been a formidable health challenge across generations, impacting 2.3 million women globally in 2022 alone. However, decades of dedicated research have gradually transformed this disease from an often fatal diagnosis to a highly treatable condition for many patients.

In the US, five-year relative survival rates now exceed 91% - a vast improvement compared to just 73.6% in 1974. Mortality rates have also seen encouraging declines, with a 41% drop in the UK since the early 1970s. In fact, despite accounting for the highest number of new cancer cases worldwide, the World Health Organization no longer ranks the disease as the leading cause of cancer deaths as of 2020.

These milestones represent the cumulative efforts of researchers, clinicians, and patients who persisted through periods of confusion and setbacks. Each new breakthrough unlocked further understanding about breast cancer's biological underpinnings and opened the door to novel therapies targeting its driving mechanisms.

3,000 BCE:

The beginnings of breast cancer research

While the word “cancer” would not appear until much later, breast cancer has been recognised for millennia, with written descriptions and illustrations that match the disease dating back to antiquity. The Edwin Smith Papyrus, an ancient Egyptian medical text from around 3,000-2,500 BCE, contains what may be the first known reference to breast cancer. 

Pictured: The Edwin Smith Papyrus. Image credit:: Jeff Dahl - Edited version of Image:EdSmPaPlateVIandVIIPrintsx.jpg,

Unlike it’s other medical papyri counterparts, which solely contained magical spells and recipes, the Edwin Smith Papyrus provides a profoundly practical guide to treating trauma in Ancient Egypt, including 48 case studies detailing traumatic injuries and treatments as observed by Egyptian physicians. Among these were detailed descriptions of several visual characteristics of “bulging” tumours of the breast. 

These firsthand accounts reveal how Egyptian physicians over 3,500 years ago grappled with diagnosing and treating this baffling disease using the limited medical knowledge of their era. Their recommended treatment was cauterisation – using a hot instrument to burn away the growths. While brutally ineffective by today's standards, it suggests these ancient doctors recognised that breast tumours required urgent intervention.

460 BCE:

Cancer in an ancient world

Across the Mediterranean – and a few millennia later – Greek physicians, including the “father of medicine” himself, Hippocrates (460 BCE), were grappling to understand the same condition that had befuddled their Egyptian predecessors. Eschewing superstitions as a cause of the disease, believing instead that cancer was a result of natural causes, Hippocrates and his disciples attributed tumours to an excess of black bile in the body, which resulted in localised growths.

Pictured: Vintage crab. Image credit: Free Public Domain Illustrations by rawpixel, licensed under CC BY 2.0.

He hypothesised that, if left untreated, the black bile tumours could spread to other bodily regions through unseen means – an astute early notion of metastasis, despite lacking the biological understanding we have today. Hippocratic physicians went on to characterise different breast tumour types in detail – from hardened, immovable masses fixed to the chest wall to ulcerated, oozing lesions – coining the term "karkinos" (Greek for crab) to describe the tumours' crab-like appearance.

This black bile theory of breast cancer's cause continued to be espoused by the later Greek physician Galen, in 200 BC, who in turn developed the Galenic humoural theory, which claimed that an excess of one of the four “humours” (phlegm, blood, yellow bile, and black bile), would disrupt the body’s natural balance, leading to illness.

16th – 18th Century:

Enter the research renaissance

In Western Europe, the study of breast cancer and medicine in general stagnated during the Middle Ages due to a variety of religious and cultural factors. Instead, many physicians during this period relied heavily on ancient Greek texts like those of Galen, taking them as authoritative and discouraging new inquiry. 

Pictured: Al Zahrawi. Image credit: Michel Bakni, CC BY-SA 4.0, via Wikimedia Commons

In contrast, the Islamic world experienced a renaissance in medicine and science during its golden age from the 8th to 13th centuries. Prominent Islamic physicians like Al-Razi (865-925 CE), Avicenna (980-1037 CE), and Al-Zahrawi (936-1013 CE) made important contributions to the detection and treatment of breast tumours. Al-Zahrawi's influential medical encyclopaedia provided one of the earliest known descriptions of mastectomy and cauterisation for breast cancer treatment.

It would not be until the 16th to 18th centuries, however, that alternative theories about breast cancer would gain significant momentum. Following the groundbreaking discovery of the lymphatic system, in the early 17th century renowned philosopher René Descartes put forward a new theory regarding the origins of breast cancer. He proposed that the disease stemmed from issues with lymphatic fluids. This "lymph theory" gained traction when the influential Scottish surgeon, John Hunter, endorsed it nearly two centuries later in the 1700s.

Hunter taught that breast tumours arose from a coagulation or curdling of defective lymph fluid. While this explanation showed little conceptual advancement from Galen's ancient notion of "black bile" causing breast cancer, it may have still encouraged surgeons of that era to more proactively remove lymph nodes in the armpit area that appeared affected during breast cancer operations.

19th century:

The golden age of surgery

Building upon the swath of medical and scientific discoveries that emerged during the renaissance, the surgical field evolved rapidly throughout the 19th century. Thanks to the introduction of radical new systems, including sterilisation, disinfection, and the use of aseptic gloves (elements that are considered baseline standards for operations today), surgery had become a safer practise for patients. 

Pictured: William Stewart Halsted. Image Credit: John H. Stocksdale - http://ihm.nlm.nih.gov/images/B14034, Public Domain

Two major breakthroughs during this period include the introduction of antiseptic surgical methods by Joseph Lister in 1867, which reduced post-operative infections, and German pathologist Rudolf Virchow's studies in cell pathology, which laid the groundwork for modern cancer research.

Against this backdrop of medical innovation, pioneering American surgeon William Halsted performed the first successful radical mastectomy (first reported in 1894), an aggressive surgical treatment for breast cancer. While radical, his approach was based on his theory that breast cancer spread primarily through the lymphatic system. By removing the breast, chest wall musculature, and lymph node chains, Halsted believed he could eliminate all sources of local and lymphatic metastasis.

Though disfiguring and intensely invasive, the radical mastectomy offered breast cancer patients a new life-extending option. As such, Halsted's technique became the standard of care for most of the 20th century, despite high rates of recurrence.

Rounding out a century moulded by innovation and discovery, after weeks of diligent research studying a new type of radiation, in 1895 German physicist Wilhelm Conrad Roentgen published his paper "On a new kind of rays". He dubbed these mysterious rays “X”.

1900s-1940s:

Radiation therapy and hormonal treatment

The latter half of the 20th century ushered in major therapeutic advances against breast cancer. Perhaps the most notable innovation to emerge during this period was chemotherapy, which was introduced as a new systemic treatment option. Drugs derived from noxious mustard gas compounds were found to combat breast tumours by disrupting rapidly dividing cancer cells.

Pictured: Charles Brenton Huggins.
Image credit: http://www.nobelprize.org/nobel_
prizes/medicine/laureates/1966/

The turn of the 20th century saw significant advancements in breast cancer research and treatment, despite the prevalence of global conflict and financial uncertainty. In 1913, researchers produced the first chemically-induced breast cancers in animal models, highlighting environmental carcinogens as risk factors.

Throughout the 1920s and 1930s, pioneering surgeons refined and promoted Halsted’s radical mastectomy techniques. A pivotal moment came in 1937 when Charles Huggins at the University of Chicago demonstrated that removing hormonal sources could block breast cancer growth, paving the way for hormone therapy. In the 1940s, radiation therapy was introduced post-mastectomy to target residual cancer cells, and modified radical mastectomies were developed, which were less disfiguring than earlier methods.

By 1949, researchers identified genetic components and inheritance patterns linked to familial breast cancer, leading to deeper exploration of DNA damage, oncogenes, and tumour suppressor genes involved in cancer development.

1950s-1960s:

Chemotherapy and improved surgical techniques

The mid-20th century brought chemotherapy into the arsenal against breast cancer. The impact of sulphur mustards on bone marrow and lymph nodes had been widely observed in soldiers during the Second World War. Noting the destructive potential of the compounds, scientists began to explore the potential of chemical-based therapies.

The initial chemotherapy drugs were derived from nitrogen mustard compounds, which were found to disrupt rapidly dividing cancer cells, including those in breast tumours.

Another breakthrough moment came in 1962, with the introduction of tamoxifen. Originally conceived (no pun intended) as Compound ICI 46,474, tamoxifen was synthesised as part of a project to develop a contraceptive pill. However, instead of supressing ovulation, as was intended, the compound in fact stimulated it. Undeterred, the research team, led by Arthur L Walpole, refocused the failed contraceptive as a way to block oestrogen receptors in some breast cancers. Patients would have to wait to access the treatment, however, as the FDA would not approve AstraZeneca’s Nolvadex (tamoxifen) until 1977.

With the chemical space transforming, it was time for surgery to get a much needed upgrade. In 1971, having spent much of his working life in the cancer research space, American surgeon Bernard Fisher led a landmark cancer trial comparing radical mastectomy with the less invasive lumpectomy – commonly known as breast conserving surgery – combined with radiotherapy. In contrast to the prevailing belief that radical mastectomy was the best course of treatment for breast cancer patients, Fisher found that the two surgical techniques had equal survival rates. Fuelled by his research, Fischer continued to call for better clinical evaluation of treatment options through the use of randomised clinical trials, and lumpectomy officially dethroned Halsted’s radical reign as the standard treatment for breast cancer.

1990s-2000s:

Genetic testing and precision medicine

The 1990s were marked by the discovery of the BRCA1 and BRCA2 genes. For decades, researchers had noted that breast cancer appeared to run in some families, suggesting hereditary factors were involved.

In 1994, after years of painstaking research mapping genetic markers, a team led by scientists at the University of Utah and National Institutes of Health finally isolated the BRCA1 (BReast CAncer 1) gene on chromosome 17. Women with inherited mutations in this gene were found to have up to an 80% lifetime risk of developing breast cancer.

Just one year later in 1995, a second gene called BRCA2 was discovered on chromosome 13 by researchers at the University of Cambridge. Harmful mutations in BRCA2 were also linked to significantly increased breast cancer risk, as well as other cancers like ovarian and prostate.

These two major gene discoveries transformed our understanding of hereditary breast cancers, which account for between 5% to 10% of all breast cancer cases. They paved the way for genetic testing and screenings to identify at-risk women with BRCA mutations.

The isolation of BRCA1/2 provided a genetic explanation for the breast cancer patterns seen in some families over generations. It also opened up new avenues for cancer prevention, such as increased screening or prophylactic mastectomies for mutation carriers.

Elsewhere, the 90s saw researchers build upon the 1974 discovery of epidermal growth factor receptor (EGFR) as a potential breast cancer target. HER2, part of the EGFR family, promotes aggressive tumour growth when overactive, a factor that had been recognised for around a decade before researchers at the biotech company Genentech revealed an innovative treatment approach – a monoclonal antibody that could specifically bind to and inhibit the effects of HER2.

This groundbreaking antibody therapy, dubbed Herceptin (trastuzumab), was shown in clinical trials to dramatically slow the progression of HER2-positive breast cancers where the HER2 oncogene was overexpressed. Given its novel mechanism of action and proven efficacy, Herceptin was granted accelerated approval by the FDA in 1998.

2001-2010:

Shedding light on the once untreatable

The dawn of the new millennium marked a paradigm shift in the way breast cancer was studied, conceptualised, and treated. Building upon the groundbreaking genetic insights of the preceding decade, the 2000s ushered in an era of personalised, molecularly-guided therapies tailored to the distinct subtypes of breast cancer.

In 2000, a landmark study published in the journal Nature used comprehensive gene expression analysis to categorise breast cancers into four intrinsic subtypes – luminal A, luminal B, HER2-enriched, and basal-like. This molecular taxonomy dramatically altered our understanding, revealing breast cancer as a heterogeneous collection of diseases with unique biological behaviours and clinical implications.

Armed with this new lens, researchers could stratify treatment strategies aligned to each subtype's molecular profile and driving oncogenic pathways. Large clinical trials in the mid-2000s validated this approach, demonstrating significant survival benefits when the HER2-targeted antibody trastuzumab was combined with chemotherapy specifically for HER2-positive disease.

As the decade progressed, other novel targeted agents, including lapatinib and bevacizumab were developed to disrupt additional growth signalling pathways in certain breast cancer subtypes. Genomic assays like Oncotype DX also emerged to predict likely chemotherapy responsiveness. However, the particularly aggressive triple-negative subtype lacking hormone receptors and HER2 overexpression remained stubbornly difficult to treat.

A pivotal milestone was reached in 2010 with the completion of the Cancer Genome Atlas' sweeping map of the entire breast cancer genomic landscape across its multiple molecular subtypes. This unprecedented molecular blueprint positioned researchers to continually refine personalised therapeutic algorithms as precision breast oncology charged forward into the next decade.

2010 - 2020:

Genomic profiling continues

Further proliferation of molecularly targeted therapies and genomic profiling strategies to combat breast cancer's heterogeneous biology continued into the 2010s. Several new targeted agents gained approval, expanding treatment options based on tumour subtype.

For HER2-positive breast cancers, additions included pertuzumab, ado-trastuzumab emtansine, and neratinib. The CDK4/6 inhibitors palbociclib, ribociclib, and abemaciclib were approved for HR-positive, HER2-negative disease, blocking cyclin-dependent kinases driving tumour growth.

Immunotherapies like the PD-L1 inhibitor atezolizumab also emerged, though their utility has initially been limited to triple-negative breast cancers with high PD-L1 expression. PARP inhibitors like olaparib and talazoparib reached the clinic for BRCA-mutated breast and ovarian cancers.

On the genomic front, next-generation sequencing and liquid biopsy technologies allowed deeper molecular profiling of breast tumours and their evolution. Multi-gene panel tests like MammaPrint, OncotypeDX, and EndoPredict were adopted to estimate recurrence risk and guide treatment.

Researchers made inroads into breast cancer heterogeneity, identifying novel molecular subtypes like luminal B HER2-enriched. They gained insights into tumour evolution, therapy resistance mechanisms, and microenvironmental interactions fuelling metastatic spread.

However, triple-negative and metastatic breast cancers remained major challenges, driving efforts to develop better predictive biomarkers and combination regimens. Building on this decade's progress, the future promised more personalised, multi-omics-informed breast cancer management.

2020-2024:

Breast cancer research in a pandemic

The COVID-19 pandemic that gripped the world in 2020 had widespread reverberations across the field of breast cancer research, diagnosis, and care delivery.

For patients already diagnosed, the pandemic created further treatment access challenges. Oncologists had to triage cases and adjust treatment regimens where possible to lower infection risks from therapies that caused immunosuppression or required frequent clinic visits. However, the use of neoadjuvant endocrine therapy increased as it allowed some ER+/HER2- breast cancer patients to delay surgery until it could be performed safely.

On the research front, many breast cancer clinical trials faced significant recruitment and enrolment challenges, with patients unable or hesitant to travel to medical centres amid the outbreak. Some trials were forced to pause enrolment or switch to remote data collection and follow-up, slowing the pace of new discoveries.

However, the pandemic also drove innovation in remote patient monitoring and catalysed efforts to improve research access and inclusion for underserved populations disproportionately impacted by both COVID-19 and breast cancer disparities. And, despite disruptions, important new treatments emerged in 2022-2023 with FDA approvals of Trodelvy for metastatic HR+ disease, Tukysa improving survival for HER2+ brain metastases, plus Enhertu and other antibody-drug conjugates, and new targeted therapies like Truqap and Orserdu for metastatic HR+ breast cancers.

While more progress remains to be made, the achievements to date solidify how sustained scientific investigation can revolutionise our ability to detect, treat, and ultimately prevent devastating diseases over time. The journey continues, fuelled by those committed to expanding the boundaries of what's possible.

About the Author

Eloise McLennan is the editor for pharmaphorum’s Deep Dive magazine. She has been a journalist and editor in the healthcare field for more than five years and has worked at several leading publications in the UK.

Sign up

Supercharge your pharma insights: Sign up to pharmaphorum's newsletter for daily updates, weekly roundups, and in-depth analysis across all industry sectors.

Click on either of the images below for more articles from this edition of Deep Dive: Oncology 2024