Immunotherapy In breast cancer

Intro to immunotherapy

  • Immunotherapy harnesses the body's own immune system to fight cancer by activating/enhancing anti-tumour immune responses.

  • It has revolutionized treatment for various cancers including breast cancer.

  • The immune system plays an important role in controlling tumour growth
    through immune surveillance.

  • However, tumours develop mechanisms to evade immune destruction forming an immunosuppressive microenvironment.

Key Immunotherapy Techniques

  • Checkpoint Inhibitors: Block inhibitory receptors (PD-1, CTLA-4) and restore anti-
    tumour T cell responses.

  • Cancer Vaccines: Stimulate adaptive immune system using tumour-associated
    antigens to develop immunological memory.

  • Adoptive Cell Therapy: Engineer/expand tumour-specific T cells outside body and
    infuse them to seek and destroy cancer cells.

  • Oncolytic Viruses: Engineered viruses that selectively replicate in and lyse tumour cells, triggering anti-tumour immunity

Mechanisms of Action

  • Checkpoint inhibitors remove brakes on immune system, allowing T cells to
    attack cancer cells directly.

  • Vaccines educate immune system to recognize and attack tumours expressing targeted antigens.

  • ACT and oncolytic viruses work alongside patient's immune cells to shrink
    established tumours.

  • Immunotherapy unleashes the power of the immune system offering new
    hope for improved treatment outcomes and long-term remissions for
    breast cancer patients.

Immunotherapy in breast cancer treatment in HER2-positive breast cancer

Role of trastuzumab

  1. HER2+ breast cancer accounts for 20% of cases and has poorer prognosis due to more aggressive disease. Trastuzumab targets this high-risk subset.

  2. Trastuzumab is a humanized monoclonal antibody immunotherapy that binds to the HER2 receptor protein.

  3. It works through dual mechanisms - inhibiting HER2 signalling and activating antibody-dependent cytotoxicity to kill cancer cells.

  4. Pivotal clinical trials demonstrated significantly improved response rates, progression-free and overall survival when trastuzumab was added to chemotherapy for HER2+ breast cancer, establishing it as a landmark targeted therapy.

Mechanism of action of trastuzumab

  • Trastuzumab binds specifically to the extracellular domain of the HER2 receptor protein. This prevents HER2 from forming heterodimers with other HER receptors such as HER1, HER3 and HER4

  • Blocking HER2 dimerization inhibits downstream PI3K-AKT and MAPK signalling pathways crucial for cell proliferation and survival

  • Trastuzumab binding also stimulates antibody-dependent cell-mediated
    cytotoxicity (ADCC) where immune cells directly kill the HER2-overexpressing cancer cells

Clinical evidence for trastuzumab in HER2+ beast cancer

• Early studies like the landmark HERceptin Adjuvant (HERA) trial showed 1-year
trastuzumab improved invasive disease-free survival by 50% compared to observation alone after adjuvant chemotherapy
5-year follow up of HERA confirmed trastuzumab provided a significant absolute risk reduction in breast cancer recurrence and mortality
Other large studies confirmed benefit when added to different chemotherapy backbones like taxanes or anthracyclines
Benefit is seen both in the neoadjuvant (before surgery) and adjuvant (after surgery) settings
Ongoing research aims to identify predictive biomarkers to stratify who benefits most and understand primary and acquired resistance

Pembrolizumab in PD-L1 expressing triple-negative breast cancer

Triple-negative breast cancer (TNBC) lacks HER2, ER and PR expression and has a poorer prognosis than other subtypes
TNBC tumours often express PD-L1 which helps the cancer evade anti-tumour
immune responses
Pembrolizumab is an anti-PD-1 monoclonal antibody immunotherapy that aims to overcome this immune evasion

Rationale for Using Pembrolizumab in PD-L1+ TNBC
PD-1 is an inhibitory receptor commonly expressed on tumour-infiltrating
lymphocytes that downregulates immunity
When its ligand PD-L1 binds to PD-1, it sends an inhibitory signal that stops T-cells from killing cancer cells
Pembrolizumab blocks this interaction, lifting the brakes on anti-tumour T-cells
within PD-L1+ TNBC tumours

Clinical Trial Results
The Phase 1b KEYNOTE-012 trial tested pembrolizumab monotherapy in patients with advanced PD-L1+ TNBC
The objective response rate was 18.5% with median response duration of 7.5 months
The responses were durable with 80% ongoing at 32-40 weeks
The managed side effects were consistent with other anti-PD-1 antibodies

The current ongoing Studies that are available are larger Phase 2 trials which assess pembrolizumab in combination with chemotherapy in advanced TNBC
Adjuvant studies are evaluating pembrolizumab after standard chemo to prevent
recurrence in early TNBC


Exploring pembrolizumab's success in treating metastatic breast cancer
Metastatic breast cancer remains largely incurable with chemotherapy and
endocrine therapies
Harnessing the immune system through checkpoint inhibitors represents a novel treatment approach
The rationale for Checkpoint Inhibition in Metastatic Breast Cancer shows that the expression of PD-L1 is seen in up to 40% of metastatic breast cancers, which acts as an immune resistance mechanism
Pembrolizumab blocks the PD-1/PD-L1 axis, releasing the brakes on anti-tumour T-cell responses
Clinical Trial Results
The multicohort KEYNOTE-012 trial evaluated pembrolizumab monotherapy in metastatic breast cancer. For PD-L1+ patients, overall response rate was 18.5% with median duration of response of 7.5 months
Responses were seen across all breast cancer subtypes regardless of hormone receptor or HER2 status
Ongoing Combination Studies include combining pembrolizumab with chemotherapy or other targeted agents may improve responses and trials that pair anti-PD-1/PD-L1 drugs with immunotherapy like tumour vaccines also
underway.


Trastuzumab Mechanism of Action
The HER2 gene is a receptor tyrosine kinase that activates growth signalling when bound by other HER ligands
In breast cancer, HER2 is often overexpressed leading to hyperactive
downstream signalling and the chief pathways among these are PI3K-AKT and MAPK, which stimulate cell proliferation and inhibits apoptosis

How Trastuzumab Works
Trastuzumab is a humanized monoclonal antibody targeted against the extracellular domain of HER2
It binds tightly to HER2 with high specificity and affinity, preventing receptor dimerization (dimerization is the process of joining two identical or similar molecular entities by bonds)
This blocks HER2 from heterodimerizing with other HER family members such as HER1, HER3, HER4
Effects on Signalling and Protein Trafficking:
By inhibiting dimerization, trastuzumab prevents downstream PI3K-AKT and MAPK cascade activation
It also induces internalization of HER2 receptors, taking them out of play at the cell surface
This decreases levels of activated signalling proteins and blocks transmission of growth signals


Immune-Mediated Effects
In addition to direct inhibition, trastuzumab activates antibody-dependent cell
cytotoxicity (ADCC) and acts as a bridge linking HER2+ cancer cells to natural killer cells of the immune system
NK cells then directly lyse and eliminate HER2-expressing breast tumour cells
coated with trastuzumab
Consequence for Breast Cancer Cells
The dual blockade of key oncogenic signalling and targeted immune killing
explains trastuzumab's potency
It inhibits HER2-driven proliferation and survival advantages conferred to breast cancer cells


Review of nivolumab's efficacy in breast cancer through targeting the PD-1 pathway
Nivolumab is a fully human IgG4 PD-1 immune checkpoint inhibitor antibody
By blocking the PD-1 receptor, nivolumab aims to reverse T-cell exhaustion and boost anti-tumour immunity
Rationale for Targeting PD-1 in Breast Cancer
PD-1 pathway activation inhibits T-cell effector function against cancer cells
Increased tumour-infiltrating lymphocytes in breast cancer express PD-1,
impairing their function
Nivolumab hopes to reinvigorate these T-cells to better attack breast
cancer cells

Clinical Trial Results
The single-arm, Phase 1/2 CheckMate-028 trial evaluated nivolumab in advanced,
refractory breast cancer
Objective response rate was 9.5% with a disease control rate of 27.6% and similar safety
to other tumours
Genomic analyses found higher ORR in patients with increased tumour mutational
burden
Ongoing Combination Studies
Trials underway adding nivolumab to chemotherapy, hormonal therapy or other
immunotherapy
Novel combinations may provide synergistic benefit and push response rates higher


Future Directions
Identifying predictive biomarkers to select optimal patients remains crucial
Earlier application as adjuvant or neoadjuvant therapy also under investigation
Nivolumab demonstrated promising initial efficacy in pretreated breast
cancer, warranting further trials
Combinatorial approaches may fully realize the potential of PD-1/PD-L1
blockade in this disease


Basic mechanism of action of immune checkpoint inhibitors
The immune system has built-in checkpoints to prevent excessive immune activation
against normal tissues
These checks include receptors like CTLA-4 and PD-1 expressed on T-cells
Cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) competes with co-stimulatory
molecule CD28
CTLA-4 binding inhibits T-cell activation and proliferation at early stages of immune
response
Ipilimumab blocks CTLA-4, removing the brakes early in an immune reaction

PD-1 Pathway Inhibition:
Programmed cell death protein 1 (PD-1) is mainly expressed by activated T-
cells
Its ligands PD-L1 and PD-L2 are often expressed on tumour and immune
cells
PD-1 ligation transmits inhibitory signals that dampen T-cell effector
function
Nivolumab/pembrolizumab disrupt this signal, preventing T-cell exhaustion

Effects on Anti-Tumour Immunity:
By inhibiting CTLA-4 and/or PD-1, immune checkpoints are released
This boosts the quantity and quality of anti-tumour T-cell responses
Primed T-cells are no longer suppressed and can better migrate, proliferate and kill
tumour cells
Clinical Impact
Tumours formerly escaping immune destruction face attacks from reinvigorated T-cells
Checkpoint blockade has revolutionized cancer treatment across various solid and liquid
tumours
Durable responses emerge from modulation of the natural anti-cancer immune capacity

Explanation of CAR-T cell therapy
What are CAR-T Cells?
Chimeric antigen receptor T (CAR-T) cells - Patient's own T
cells engineered with a chimeric antigen receptor
How CAR-T Cells Work
T cells are collected from a patient through leukapheresis then the genes for an artificial CAR are inserted
The CAR allows T cells to recognise a specific antigen on tumour cells independent of MHC
Once reinfused, CAR-T cells can target and kill cancer cells expressing the antigen

Components of CAR
An extracellular domain anchors to the target antigen
A transmembrane domain anchors the receptor to the T cell
An intracellular domain provides co-stimulation and activates T cell functions upon binding
target
Significance in Cancer Immunotherapy
CAR-T cells redirect the potent killing ability of T cells to eliminate cancers
They show dramatic responses in some hematologic cancers resistant to other therapies
Potential to treat both liquid and solid tumours that express unique antigens
Explanation of CAR-T cell therapy

Challenges and Applications
Safety concerns include cytokine release syndrome and neurotoxicity from high activation
Identifying targets specific to cancer cells vs normal tissues is crucial
Ongoing research optimizes design, targets, and combination with other agents
CAR-T cell therapy harnesses the immune system's precision to provide a potentially curative approach
While still developing, it represents a breakthrough in immunotherapy that may transform cancer treatment

Evaluation of the primary role of monoclonal antibodies in cancer immunotherapy focusing on their mechanism of action
Role of Monoclonal Antibodies
Monoclonal antibodies are engineered versions of immune system proteins able to precisely target molecular markers on cancer cells
Mechanisms of Action
Direct Blockade - Binding to targets like HER2 receptors interferes with downstream oncogenic signalling
Opsonization - Coating tumour cells allows for antibody-dependent cellular cytotoxicity via NK cells and macrophages
Immune Cell Recruitment - Effector functions stimulate an inflammatory response and attract immune cells to the tumour
Co-stimulation - Increasing supportive signals or blocking inhibitory receptors licenses T cell anti-tumour activity

Key Applications in Cancer Immunotherapy
Delivery of Radioisotopes/Chemotherapy - Conjugating agents directly delivers drugs to cancer tissue sparing healthy cells
Checkpoint Inhibition - Anti-CTLA-4, PD-1, PD-L1 antibodies harness pre-existing immunity by removing brakes from T cells
Targeted Delivery - CAR-T cells armed with tumour antigen recognition via recombinant monoclonal antibodies
Benefits
Highly specific targeting limits off-tumour toxicities compared to traditional therapies
Persistence allows for durable clinical responses through sustained immune modulation
Combination with other modalities increases potency by attacking tumours through multiple avenues

The primary goal of immune checkpoint inhibitors in cancer treatment is to restore and enhance the body's natural anti-tumour immune response
Cancers have developed mechanisms to evade immune destruction, such as overexpressing immune checkpoint proteins like CTLA-4 and PD-1.
These checkpoint proteins act as "brakes" that inhibit T cell activation and antitumor activity when they bind to ligands on tumours cells.
Checkpoint inhibitors are monoclonal antibodies that block these inhibitory checkpoint interactions between T cells and cancer cells.
By disrupting checkpoints, the goal is to "release the brakes" on T cells and empower them to better recognise tumours as foreign threats.
• Uninhibited by checkpoint signals, T cells can proliferate robustly at tumour sites and launch

more effective attacks on cancer via their cytotoxic and cytokine releasing functions.
This aims to turn a "cold" immunosuppressive tumour microenvironment back into a "hot" inflamed one that drives a potent immunotherapeutic response.
The ultimate goal is durable regression or even cure of cancers by restoring and enhancing the body's natural antitumor defences, rather than directly killing cancer cells like chemotherapy.
By unleashing pre-existing anti-tumour immunity, checkpoint inhibitors offer a potentially self-sustaining treatment paradigm.
The overarching objective is reinvigorating and optimizing the immune system's intrinsic ability to eliminate cancers on its own.

Explanation of tumour-infiltrating
lymphocyte (TIL) therapy
TIL therapy involves extracting T cells from a patient's own tumour tissue that are actively
infiltrating the tumour microenvironment.
These TILs have already demonstrated an ability to recognise tumour antigens and traffic to
tumour sites.
The TILs are then expanded ex vivo by activating and stimulating them with IL-2 and anti-
CD3/CD28 antibodies in culture over 2-4 weeks.
This amplification process increases the number of TILs, especially those specifically
targeting the patient's tumour.
The expanded population of tumour-reactive TILs are then infused back into the patient to
induce an antitumour immune response.
TIL therapy has shown durable responses in metastatic melanoma when combined with
lymphodepletion to remove regulatory T cells.

It aims to harness the immune system's intrinsic ability to generate tumour-specific T cell
clones within tumours.
Identifying novel tumour antigens recognised by TILs also helps develop more targeted
immunotherapies.
Current research optimises TIL culture methods and explores combining with checkpoint
inhibitors or adoptive NK cell therapy.
TIL therapy isolates, amplifies and redeploys a patient's pre-existing tumour-specific T
cells to induce potent, personalised antitumour immunity.