BSI Immunology

BiteSized Immunology: Systems & Processes

Bitesized Immunology

Antigen Processing and Presentation

In order to be capable of engaging the key elements of adaptive immunity (specificity, memory, diversity, self/nonself discrimination), antigens have to be processed and presented to immune cells. Antigen presentation is mediated by MHC class I molecules , and the class II molecules found on the surface of antigen-presenting cells (APCs) and certain other cells.

MHC class I and class II molecules are similar in function: they deliver short peptides to the cell surface allowing these peptides to be recognised by CD8+ (cytotoxic) and CD4+ (helper) T cells, respectively. The difference is that the peptides originate from different sources – endogenous, or intracellular , for MHC class I; and exogenous, or extracellular for MHC class II. There is also so called cross-presentation in which exogenous antigens can be presented by MHC class I molecules. Endogenous antigens can also be presented by MHC class II when they are degraded through autophagy.

Antigen processing and presentation

MHC class I presentation

MHC class I molecules are expressed by all nucleated cells. MHC class I molecules are assembled in the endoplasmic reticulum (ER) and consist of two types of chain – a polymorphic heavy chain and a chain called β2-microglobulin. The heavy chain is stabilised by the chaperone calnexin , prior to association with the β2-microglobulin. Without peptides, these molecules are stabilised by chaperone proteins : calreticulin, Erp57, protein disulfide isomerase (PDI) and tapasin. The complex of TAP, tapasin, MHC class I, ERp57 and calreticulin is called the peptide-loading complex (PLC). Tapasin interacts with the transport protein TAP (transporter associated with antigen presentation) which translocates peptides from the cytoplasm into the ER. Prior to entering the ER, peptides are derived from the degradation of proteins, which can be of viral- or self origin. Degradation of proteins is mediated by cytosolic- and nuclear proteasomes, and the resulting peptides are translocated into the ER by means of TAP. TAP translocates peptides of 8 –16 amino acids and they may require additional trimming in the ER before binding to MHC class I molecules. This is possibly due to the presence of ER aminopeptidase (ERAAP) associated with antigen processing.

It should be noted that 30–70% of proteins are immediately degraded after synthesis (they are called DRiPs – defective ribosomal products, and they are the result of defective transcription or translation). This process allows viral peptides to be presented very quickly – for example, influenza virus can be recognised by T cells approximately 1.5 hours post-infection. When peptides bind to MHC class I molecules, the chaperones are released and peptide–MHC class I complexes leave the ER for presentation at the cell surface. In some cases, peptides fail to associate with MHC class I and they have to be returned to the cytosol for degradation. Some MHC class I molecules never bind peptides and they are also degraded by the ER-associated protein degradation (ERAD) system.

There are different proteasomes that generate peptides for MHC class-I presentation: 26S proteasome , which is expressed by most cells; the immunoproteasome, which is expressed by many immune cells; and the thymic-specific proteasome expressed by thymic epithelial cells.

Antigen presentation

On the surface of a single cell, MHC class I molecules provide a readout of the expression level of up to 10,000 proteins. This array is interpreted by cytotoxic T lymphocytes and Natural Killer cells, allowing them to monitor the events inside the cell and detect infection and tumorigenesis.

MHC class I complexes at the cell surface may dissociate as time passes and the heavy chain can be internalised. When MHC class I molecules are internalised into the endosome, they enter the MHC class-II presentation pathway. Some of the MHC class I molecules can be recycled and present endosomal peptides as a part of a process which is called cross-presentation .

The usual process of antigen presentation through the MHC I molecule is based on an interaction between the T-cell receptor and a peptide bound to the MHC class I molecule. There is also an interaction between the CD8+ molecule on the surface of the T cell and non-peptide binding regions on the MHC class I molecule. Thus, peptide presented in complex with MHC class I can only be recognised by CD8+ T cells. This interaction is a part of so-called ‘three-signal activation model’, and actually represents the first signal. The next signal is the interaction between CD80/86 on the APC and CD28 on the surface of the T cell, followed by a third signal – the production of cytokines by the APC which fully activates the T cell to provide a specific response.

MHC class I polymorphism

Human MHC class I molecules are encoded by a series of genes – HLA-A, HLA-B and HLA-C (HLA stands for ‘Human Leukocyte Antigen’, which is the human equivalent of MHC molecules found in most vertebrates). These genes are highly polymorphic, which means that each individual has his/her own HLA allele set. The consequences of these polymorphisms are differential susceptibilities to infection and autoimmune diseases that may result from the high diversity of peptides that can bind to MHC class I in different individuals. Also, MHC class I polymorphisms make it virtually impossible to have a perfect tissue match between donor and recipient, and thus are responsible for graft rejection.

Antigen processing and presentation

MHC class II presentation

MHC class II molecules are expressed by APCs, such as dendritic cells (DC), macrophages and B cells (and, under IFNγ stimuli, by mesenchymal stromal cells, fibroblasts and endothelial cells, as well as by epithelial cells and enteric glial cells). MHC class II molecules bind to peptides that are derived from proteins degraded in the endocytic pathway. MHC class II complexes consists of α- and β-chains that are assembled in the ER and are stabilised by invariant chain (Ii). The complex of MHC class II and Ii is transported through the Golgi into a compartment which is termed the MHC class II compartment (MIIC). Due to acidic pH, proteases cathepsin S and cathepsin L are activated and digest Ii, leaving a residual class II-associated Ii peptide (CLIP) in the peptide-binding groove of the MHC class II. Later, the CLIP is exchanged for an antigenic peptide derived from a protein degraded in the endosomal pathway. This process requires the chaperone HLA-DM, and, in the case of B cells, the HLA-DO molecule. MHC class II molecules loaded with foreign peptide are then transported to the cell membrane to present their cargo to CD4+ T cells. Thereafter, the process of antigen presentation by means of MHC class II molecules basically follows the same pattern as for MHC class I presentation.

As opposed to MHC class I, MHC class II molecules do not dissociate at the plasma membrane. The mechanisms that control MHC class II degradation have not been established yet, but MHC class II molecules can be ubiquitinised and then internalised in an endocytic pathway.

MHC class II polymorphism

Like the MHC class I heavy chain, human MHC class II molecules are encoded by three polymorphic genes: HLA-DR, HLA-DQ and HLA-DP. Different MHC class II alleles can be used as genetic markers for several autoimmune diseases, possibly owing to the peptides that they present.

explain the process of antigen presentation

  • DNA Replication
  • Active Transport
  • Cellular Receptors
  • Endocytosis and Exocytosis
  • Enzyme Inhibition
  • Enzyme Kinetics
  • Protein Structure
  • Transcription of DNA
  • Translation of DNA
  • Anaerobic Respiration
  • Electron Transport Chain
  • Gluconeogenesis
  • Calcium Regulation
  • External Balance of Potassium
  • Internal Balance of Potassium
  • Sodium Regulation
  • Cell Membrane
  • Endoplasmic Reticulum
  • Golgi Apparatus
  • Mitochondria
  • Blood Vessels
  • Cellular Adaptations
  • Epithelial Cells
  • Muscle Histology
  • Structure of Glands
  • Control of Stroke Volume
  • Control of Heart Rate
  • Cardiac Cycle
  • Cardiac Pacemaker Cells
  • Conduction System
  • Contraction of Cardiac Muscle
  • Ventricular Action Potentials
  • Blood Flow in Vessels
  • Control of Blood Pressure
  • Capillary Exchange
  • Flow In Peripheral Circulation
  • Venous Return
  • Cardiac Muscle
  • Hepatic Circulation
  • Skeletal Muscle
  • Airway Resistance
  • Lung Volumes
  • Mechanics of Breathing
  • Gas Exchange
  • Oxygen Transport in The Blood
  • Transport of Carbon Dioxide in the Blood
  • Ventilation-Perfusion Matching
  • Chemoreceptors
  • Cough Reflex
  • Neural Control of Ventilation
  • Respiratory Regulation of Acid-Base Balance
  • Responses of The Respiratory System to Stress
  • Regulation of Saliva
  • Secretion of Saliva
  • Gastric Acid Production
  • Gastric Mucus Production
  • Digestion and Absorption
  • Histology and Cellular Function of the Small Intestine
  • Absorption in the Large Intestine
  • Large Intestinal Motility
  • Bilirubin Metabolism
  • Carbohydrate Metabolism in the Liver
  • Lipid Metabolism in the Liver
  • Protein and Ammonia Metabolism in the Liver
  • Storage Functions of the Liver
  • Bile Production
  • Function of The Spleen
  • Exocrine Pancreas
  • Somatostatin
  • Proximal Convoluted Tubule
  • Loop of Henle
  • Distal Convoluted Tubule and Collecting Duct
  • Storage Phase of Micturition
  • Voiding Phase of Micturition
  • Antidiuretic Hormone
  • Renin-Angiotensin-Aldosterone System
  • Urinary Regulation of Acid-Base Balance
  • Water Filtration and Reabsorption
  • Development of the Reproductive System
  • Gametogenesis
  • Gonadotropins and the Hypothalamic Pituitary Axis
  • Menstrual Cycle
  • Placental Development
  • Fetal Circulation
  • Maternal Adaptations in Pregnancy
  • Cells of the Nervous System
  • Central Nervous System
  • Cerebrospinal Fluid
  • Neurotransmitters
  • Peripheral Nervous System
  • Action Potential
  • Excitatory and Inhibitory Synaptic Signalling
  • Resting Membrane Potential
  • Synaptic Plasticity
  • Synaptic Transmission
  • Ascending Tracts
  • Auditory Pathway
  • Consciousness and Sleep
  • Modalities of Sensation
  • Pain Pathways
  • Sensory Acuity
  • Visual Pathway
  • Descending Tracts
  • Lower Motor Neurones
  • Muscle Stretch Reflex
  • Upper Motor Neurones
  • Aqueous Humour
  • Ocular Accommodation
  • Thyroid Gland
  • Parathyroid Glands
  • Adrenal Medulla
  • Zona Glomerulosa
  • Zona Fasciculata
  • Zona Reticularis
  • Endocrine Pancreas
  • The Hypothalamus
  • Anterior Pituitary
  • Posterior Pituitary
  • White Blood Cells – Summary
  • Barriers to Infection
  • Infection Recognition Molecules
  • Phagocytosis
  • The Complement System

Antigen Processing and Presentation

  • Primary and Secondary Immune Responses
  • T Cell Memory
  • Acute Inflammation
  • Autoimmunity
  • Chronic Inflammation
  • Hypersensitivity Reactions
  • Immunodeficiency
  • Types of Immunity
  • Antibiotics
  • Viral Infection
  • Blood Groups
  • Coagulation
  • Erythropoiesis
  • Iron Metabolism
  • Mononuclear Phagocyte System

Original Author(s): Antonia Round Last updated: 17th July 2023 Revisions: 9

  • 1 Antigen Presentation
  • 2.1 MHC Class I Molecules
  • 2.2 MCH Class II Molecules
  • 3.1 T Cell Receptors
  • 3.2 Co-Receptors
  • 4 Clinical Relevance – Autoimmune disease

T cells can only recognise antigens when they are displayed on cell surfaces. This is carried out by  Antigen-presenting cells (APCs) , the most important of which are dendritic cells, B cells, and macrophages. APCs can digest proteins they encounter and display peptide fragments from them on their surfaces for other immune cells to recognise.

This process of antigen presentation allows T cells to “see” what proteins are present in the body and to form an adaptive immune response against them. In this article, we shall discuss antigen processing, presentation, and recognition by T cells.

Antigen Presentation

Antigens are delivered to the surface of APCs by Major Histocompatibility Complex (MHC) molecules. Different MHC molecules can bind different peptides. The MHC is highly polygenic and polymorphic which equips us to recognise a vast array of different antigens we might encounter. There are different classes of MHC, which have different functions:

  • MHC class I  molecules are found on all nucleated cells (not just professional APCs) and typically present intracellular antigens such as viruses.
  • MHC class II molecules are only found on APCs and typically present extracellular antigens such as bacteria.

This is logical because should a virus be inside a cell of any type, the immune system needs to be able to respond to it. This also explains why pathogens inside human red blood cells (which are non-nucleated) can be difficult for the immune system to find, such as in malaria.

Whilst this is the general rule, in cross-presentation extracellular antigens can be presented by MHC class I, and in autophagy intracellular antigens can be presented by MHC class II.

Antigen Processing

Before an antigen can be presented, it must first be processed . Processing transforms proteins into antigenic peptides.

MHC Class I Molecules

Intracellular peptides for MHC class I presentation are made by proteases and the proteasome in the cytosol, then transported into the endoplasmic reticulum via TAP (Transporter associated with Antigen Processing) to be further processed.

They are then assembled together with MHC I molecules and travel to the cell surface ready for presentation.

explain the process of antigen presentation

Fig 1 – Diagram demonstrating the production of peptides for MHC class I presentation

MCH Class II Molecules

The route of processing for exogenous antigens for MHC class II presentation begins with endocytosis of the antigen. Once inside the cell, they are encased within endosomes that acidify and activate proteases, to degrade the antigen.

MHC class II molecules are transported into endocytic vesicles where they bind peptide antigen and then travel to the cell surface.

explain the process of antigen presentation

Fig 2 – Diagram showing processing of antigens for MHC Class II presentation by a dendritic cell

The antigen presented on MHCs is recognised by T cells using a T cell receptor (TCR) . These are  antigen-specific .

T Cell Receptors

Each T cell has thousands of TCRs , each with a unique specificity that collectively allows our immune system to recognise a wide array of antigens.

This diversity in TCRs is achieved through a process called V(D)J recombination during development in the thymus. TCR chains have a variable region where gene segments are randomly rearranged, using the proteins RAG1 and RAG2 to initiate cleavage and non-homologous end joining to rejoin the chains.

The diversity of the TCRs can be further increased by inserting or deleting nucleotides at the junctions of gene segments; together forming the potential to create up to 10 15 unique TCRs.

TCRs are specific not only for a particular antigen but also for a specific MHC molecule. T cells will only recognise an antigen if a specific antigen with a specific MHC molecule is present: this phenomenon is called  MHC restriction .

Co-Receptors

As well as the TCR, another T cell molecule is required for antigen recognition and is known as a co-receptor. These are either a CD4 or CD8 molecule:

  • CD4 is present on T helper cells and only binds to antigen-MHC II complexes.
  • CD8 is present on cytotoxic T cells and only binds to antigen-MHC I complexes.

This, therefore, leads to very different effects. Antigens presented with MHC II will activate T helper cells and antigens presented with MHC I activate cytotoxic T cells. Cytotoxic T cells will kill the cells that they recognise, whereas T helper cells have a broader range of effects on the presenting cell such as activation to produce antibodies (in the case of B cells) or activation of macrophages to kill their intracellular pathogens.

Clinical Relevance – Autoimmune disease

It is important to note that APCs may deliver foreign antigens or self-antigens. In the case of autoimmune diseases, self-antigens are presented to T cells, which then initiates an immune response against our own tissues.

For example, in Graves’ disease , TSHR (thyroid stimulating hormone receptor) acts as a self-antigen and is presented to T cells. This then activates B cells to produce autoantibodies against TSHRs in the thyroid. This results in the activation of TSHRs leading to hyperthyroidism and a possible goitre.

[start-clinical]

Clinical Relevance - Autoimmune disease

[end-clinical]

Found an error? Is our article missing some key information? Make the changes yourself here!

Once you've finished editing, click 'Submit for Review', and your changes will be reviewed by our team before publishing on the site.

We use cookies to improve your experience on our site and to show you relevant advertising. To find out more, read our privacy policy .

Privacy Overview

Library homepage

  • school Campus Bookshelves
  • menu_book Bookshelves
  • perm_media Learning Objects
  • login Login
  • how_to_reg Request Instructor Account
  • hub Instructor Commons

Margin Size

  • Download Page (PDF)
  • Download Full Book (PDF)
  • Periodic Table
  • Physics Constants
  • Scientific Calculator
  • Reference & Cite
  • Tools expand_more
  • Readability

selected template will load here

This action is not available.

Medicine LibreTexts

20.3E: Antigen-Presenting Cells

  • Last updated
  • Save as PDF
  • Page ID 7949

\( \newcommand{\vecs}[1]{\overset { \scriptstyle \rightharpoonup} {\mathbf{#1}} } \)

\( \newcommand{\vecd}[1]{\overset{-\!-\!\rightharpoonup}{\vphantom{a}\smash {#1}}} \)

\( \newcommand{\id}{\mathrm{id}}\) \( \newcommand{\Span}{\mathrm{span}}\)

( \newcommand{\kernel}{\mathrm{null}\,}\) \( \newcommand{\range}{\mathrm{range}\,}\)

\( \newcommand{\RealPart}{\mathrm{Re}}\) \( \newcommand{\ImaginaryPart}{\mathrm{Im}}\)

\( \newcommand{\Argument}{\mathrm{Arg}}\) \( \newcommand{\norm}[1]{\| #1 \|}\)

\( \newcommand{\inner}[2]{\langle #1, #2 \rangle}\)

\( \newcommand{\Span}{\mathrm{span}}\)

\( \newcommand{\id}{\mathrm{id}}\)

\( \newcommand{\kernel}{\mathrm{null}\,}\)

\( \newcommand{\range}{\mathrm{range}\,}\)

\( \newcommand{\RealPart}{\mathrm{Re}}\)

\( \newcommand{\ImaginaryPart}{\mathrm{Im}}\)

\( \newcommand{\Argument}{\mathrm{Arg}}\)

\( \newcommand{\norm}[1]{\| #1 \|}\)

\( \newcommand{\Span}{\mathrm{span}}\) \( \newcommand{\AA}{\unicode[.8,0]{x212B}}\)

\( \newcommand{\vectorA}[1]{\vec{#1}}      % arrow\)

\( \newcommand{\vectorAt}[1]{\vec{\text{#1}}}      % arrow\)

\( \newcommand{\vectorB}[1]{\overset { \scriptstyle \rightharpoonup} {\mathbf{#1}} } \)

\( \newcommand{\vectorC}[1]{\textbf{#1}} \)

\( \newcommand{\vectorD}[1]{\overrightarrow{#1}} \)

\( \newcommand{\vectorDt}[1]{\overrightarrow{\text{#1}}} \)

\( \newcommand{\vectE}[1]{\overset{-\!-\!\rightharpoonup}{\vphantom{a}\smash{\mathbf {#1}}}} \)

Antigen presentation is a process by which immune cells capture antigens and then enable their recognition by T cells.

Learning Objectives

  • Describe the role of antigen-presenting cells
  • The host’s cells express “self” antigens that identify them as such. These antigens are different from those in bacteria (“non-self” antigens) and in virus-infected host cells (“missing-self”).
  • Antigen presentation consists of pathogen recognition, phagocytosis of the pathogen or its molecular components, processing of the antigen, and then presentation of the antigen to naive T cells.
  • The T cell receptor is restricted to recognizing antigenic peptides only when bound to appropriate molecules of the major histocompatibility complex (MHC), also known in humans as human leukocyte antigen (HLA).
  • Helper T cells recieve antigens from MHC II on an APC, while cytotoxic T cells recieve antigens from MHC I. Helper T cells present their antigen to B cells as well.Dendritic cells, B cells, and macrophages play a major role in the innate response, and are the primary antigen-presenting cells (APC).
  • APCs use toll-like receptors to identify PAMPS and DAMPs, which are signs of an infection and may be processed into antigen peptides if phagocytized. Most APCs cannot tell the difference between different types of antigens like B and T cells can.
  • damage-associated molecular pattern : Protein or nucleic acid based signs of pathogen induced damage. Protein DAMPs may be phagocytized and processed for antigen presentation.
  • cytotoxic : A population of T cells specialized for inducing the deaths of other cells.

Antigen presentation is a process in the body’s immune system by which macrophages, dendritic cells and other cell types capture antigens, then present them to naive T-cells. The basis of adaptive immunity lies in the capacity of immune cells to distinguish between the body’s own cells and infectious pathogens. The host’s cells express “self” antigens that identify them as belonging to the self. These antigens are different from those in bacteria (“non-self” antigens) or in virally-infected host cells (“missing-self”). Antigen presentation broadly consists of pathogen recognition, phagocytosis of the pathogen or its molecular components, processing of the antigen, and then presentation of the antigen to naive (mature but not yet activated) T cells. The ability of the adaptive immune system to fight off pathogens and end an infection depends on antigen presentation.

Antigen Presenting Cells

Antigen Presenting Cells (APCs) are cells that capture antigens from within the body, and present them to naive T-cells. Many immune system cells can present antigens, but the most common types are macrophages and dendritic cells, which are two types of terminally differentiated leukocytes that arise from monocytes. Both of these APCs perform many immune functions that are important for both innate and adaptive immunity, such as removing leftover pathogens and dead neutrophils after an inflammatory response. Dendritic cells (DCs) are generally found in tissues that have contact with the external environment (such as the skin or respiratory epithelium) while macrophages are found in almost all tissues. Some types of B cells may also present antigens as well, though it is not their primary function.

APCs phagocytize exogenous pathogens such as bacteria, parasites, and toxins in the tissues and then migrate, via chemokine signals, to lymph nodes that contain naive T cells. During migration, APCs undergo a process of maturation in which they digest phagocytized pathogens and begin to express the antigen in the form of a peptide on their MHC complexes, which enables them to present the antigen to naive T cells. The antigen digestion phase is also called “antigen processing,” because it prepares the antigens for presentation. This MHC:antigen complex is then recognized by T cells passing through the lymph node. Exogenous antigens are usually displayed on MHC Class II molecules, which interact with CD4+ helper T cells.

This maturation process is dependent on signaling from other pathogen-associated molecular pattern (PAMP) molecules (such as a toxin or component of a cell membrane from a pathogen) through pattern recognition receptors (PRRs), which are received by Toll-like receptors on the DC’s body. They may also recognize damage-associated molecular pattern (DAMP) molecules, which include degraded proteins or nucleic acids released from cells that undergo necrosis. PAMPs and DAMPS are not technically considered antigens themselves, but instead are signs of pathogen presence that alert APCs through Toll-like receptor binding. However if a DC phagocytzes a PAMP or DAMP, it could be used as an antigen during antigen presentation. APCs are unable to distinguish between different types of antigens themselves, but B and T cells can due to their specificity.

Antigen Presentation

T cells must be presented with antigens in order to perform immune system functions. The T cell receptor is restricted to recognizing antigenic peptides only when bound to appropriate molecules of the MHC complexes on APCs, also known in humans as Human leukocyte antigen (HLA).

Several different types of T cell can be activated by APCs, and each type of T cell is specially equipped to deal with different pathogens, whether the pathogen is bacterial, viral or a toxin. The type of T cell activated, and therefore the type of response generated, depends on which MHC complex the processed antigen-peptide binds to.

MHC Class I molecules present antigen to CD8+ cytotoxic T cells, while MHC class II molecules present antigen to CD4+ helper T cells. With the exception of some cell types (such as erythrocytes), Class I MHC is expressed by almost all host cells. Cytotoxic T cells (also known as TC, killer T cell, or cytotoxic T-lymphocyte (CTL)) are a population of T cells that are specialized for inducing the death of other cells. Recognition of antigenic peptides through Class I by CTLs leads to the killing of the target cell, which is infected by virus, intracytoplasmic bacterium, or are otherwise damaged or dysfunctional. Additionally, some helper T cells will present their antigen to B cells, which will activate their proliferation response.

Antigen presentation : In the upper pathway; foreign protein or antigen (1) is taken up by an antigen-presenting cell (2). The antigen is processed and displayed on a MHC II molecule (3), which interacts with a T helper cell (4). In the lower pathway; whole foreign proteins are bound by membrane antibodies (5) and presented to B lymphocytes (6), which process (7) and present antigen on MHC II (8) to a previously activated T helper cell (10), spurring the production of antigen-specific antibodies (9).

LICENSES AND ATTRIBUTIONS

CC LICENSED CONTENT, SHARED PREVIOUSLY

  • Curation and Revision. Authored by : Boundless.com. Provided by : Boundless.com. License : CC BY-SA: Attribution-ShareAlike

CC LICENSED CONTENT, SPECIFIC ATTRIBUTION

  • antigen. Provided by : Wiktionary. Located at : en.wiktionary.org/wiki/antigen . License : CC BY-SA: Attribution-ShareAlike
  • Adaptive immune system. Provided by : Wikipedia. Located at : en.Wikipedia.org/wiki/Adaptive_immune_system . License : CC BY-SA: Attribution-ShareAlike
  • antibody. Provided by : Wiktionary. Located at : en.wiktionary.org/wiki/antibody . License : CC BY-SA: Attribution-ShareAlike
  • macrophage. Provided by : Wiktionary. Located at : en.wiktionary.org/wiki/macrophage . License : CC BY-SA: Attribution-ShareAlike
  • Antibody. Provided by : Wikipedia. Located at : en.Wikipedia.org/wiki/File:Antibody.jpg . License : Public Domain: No Known Copyright
  • Antigen presentation. Provided by : Wikipedia. Located at : en.Wikipedia.org/wiki/File:Antigen_presentation.svg . License : Public Domain: No Known Copyright
  • Immune system. Provided by : Wikipedia. Located at : en.Wikipedia.org/wiki/Immune_system%23Innate_immune_system . License : CC BY-SA: Attribution-ShareAlike
  • Lymphocyte_activation_simple.png. Provided by : Wikipedia. Located at : en.Wikipedia.org/wiki/Immune_system#/media/File:Lymphocyte_activation_simple.png . License : CC BY-SA: Attribution-ShareAlike
  • T cell. Provided by : Wikipedia. Located at : en.Wikipedia.org/wiki/T_cell . License : CC BY-SA: Attribution-ShareAlike
  • B cell. Provided by : Wikipedia. Located at : en.Wikipedia.org/wiki/B_cell . License : CC BY-SA: Attribution-ShareAlike
  • Immune cells. Provided by : Wikipedia. Located at : en.Wikipedia.org/wiki/Immune_cells . License : CC BY-SA: Attribution-ShareAlike
  • thymus. Provided by : Wiktionary. Located at : en.wiktionary.org/wiki/thymus . License : CC BY-SA: Attribution-ShareAlike
  • Red White Blood cells. Provided by : Wikipedia. Located at : en.Wikipedia.org/wiki/File:Red_White_Blood_cells.jpg . License : Public Domain: No Known Copyright
  • Lymphocyte. Provided by : Wikipedia. Located at : en.Wikipedia.org/wiki/Lymphocyte . License : CC BY-SA: Attribution-ShareAlike
  • B cells. Provided by : Wikipedia. Located at : en.Wikipedia.org/wiki/B%20cells . License : CC BY-SA: Attribution-ShareAlike
  • natural killer (NK) cells. Provided by : Wikipedia. Located at : en.Wikipedia.org/wiki/natural...20(NK)%20cells . License : CC BY-SA: Attribution-ShareAlike
  • T cells. Provided by : Wiktionary. Located at : en.wiktionary.org/wiki/T_cells . License : CC BY-SA: Attribution-ShareAlike
  • Lymphocyte. Provided by : Wikipedia. Located at : en.Wikipedia.org/wiki/Lymphocyte . License : Public Domain: No Known Copyright
  • Antigen presentation. Provided by : Wikipedia. Located at : en.Wikipedia.org/wiki/Antigen_presentation . License : CC BY-SA: Attribution-ShareAlike
  • cytotoxic. Provided by : Wiktionary. Located at : en.wiktionary.org/wiki/cytotoxic . License : CC BY-SA: Attribution-ShareAlike
  • Antigen presentation. Provided by : Wikipedia. Located at : en.Wikipedia.org/wiki/Antigen_presentation . License : Public Domain: No Known Copyright

Module 20: The Immune System

Antigen-presenting cells, learning outcomes.

  • Describe the structure and function of antigen-presenting cells

Unlike NK cells of the innate immune system, B cells (B lymphocytes) are a type of white blood cell that gives rise to antibodies, whereas T cells (T lymphocytes) are a type of white blood cell that plays an important role in the immune response. T cells are a key component in the cell-mediated response—the specific immune response that utilizes T cells to neutralize cells that have been infected with viruses and certain bacteria. There are three types of T cells: cytotoxic, helper, and suppressor T cells. Cytotoxic T cells destroy virus-infected cells in the cell-mediated immune response, and helper T cells play a part in activating both the antibody and the cell-mediated immune responses. Suppressor T cells deactivate T cells and B cells when needed, and thus prevent the immune response from becoming too intense.

An antigen is a foreign or “non-self” macromolecule that reacts with cells of the immune system. Not all antigens will provoke a response. For instance, individuals produce innumerable “self” antigens and are constantly exposed to harmless foreign antigens, such as food proteins, pollen, or dust components. The suppression of immune responses to harmless macromolecules is highly regulated and typically prevents processes that could be damaging to the host, known as tolerance.

The innate immune system contains cells that detect potentially harmful antigens, and then inform the adaptive immune response about the presence of these antigens. An antigen-presenting cell (APC) is an immune cell that detects, engulfs, and informs the adaptive immune response about an infection. When a pathogen is detected, these APCs will phagocytose the pathogen and digest it to form many different fragments of the antigen. Antigen fragments will then be transported to the surface of the APC, where they will serve as an indicator to other immune cells. Dendritic cells are immune cells that process antigen material; they are present in the skin (Langerhans cells) and the lining of the nose, lungs, stomach, and intestines. Sometimes a dendritic cell presents on the surface of other cells to induce an immune response, thus functioning as an antigen-presenting cell. Macrophages also function as APCs. Before activation and differentiation, B cells can also function as APCs.

After phagocytosis by APCs, the phagocytic vesicle fuses with an intracellular lysosome forming phagolysosome. Within the phagolysosome, the components are broken down into fragments; the fragments are then loaded onto MHC class I or MHC class II molecules and are transported to the cell surface for antigen presentation, as illustrated in Figure 1. Note that T lymphocytes cannot properly respond to the antigen unless it is processed and embedded in an MHC II molecule. APCs express MHC on their surfaces, and when combined with a foreign antigen, these complexes signal a “non-self” invader. Once the fragment of antigen is embedded in the MHC II molecule, the immune cell can respond. Helper T- cells are one of the main lymphocytes that respond to antigen-presenting cells. Recall that all other nucleated cells of the body expressed MHC I molecules, which signal “healthy” or “normal.”

Illustration shows a bacterium being engulfed by a macrophage. Lysosomes fuse with the vacuole containing the bacteria. The bacterium is digested. Antigens from the bacterium are attached to a MHC II molecule and presented on the cell surface.

Figure 1. An APC, such as a macrophage, engulfs and digests a foreign bacterium. An antigen from the bacterium is presented on the cell surface in conjunction with an MHC II molecule Lymphocytes of the adaptive immune response interact with antigen-embedded MHC II molecules to mature into functional immune cells.

  • Biology 2e. Provided by : OpenStax. Located at : http://cnx.org/contents/[email protected] . License : CC BY: Attribution . License Terms : Access for free at https://openstax.org/books/biology-2e/pages/1-introduction

Footer Logo Lumen Waymaker

Library homepage

  • school Campus Bookshelves
  • menu_book Bookshelves
  • perm_media Learning Objects
  • login Login
  • how_to_reg Request Instructor Account
  • hub Instructor Commons

Margin Size

  • Download Page (PDF)
  • Download Full Book (PDF)
  • Periodic Table
  • Physics Constants
  • Scientific Calculator
  • Reference & Cite
  • Tools expand_more
  • Readability

selected template will load here

This action is not available.

Biology LibreTexts

12.3B: Antigen-Presenting Cells (APCs)

  • Last updated
  • Save as PDF
  • Page ID 3295

  • Gary Kaiser
  • Community College of Baltimore Country (Cantonsville)

\( \newcommand{\vecs}[1]{\overset { \scriptstyle \rightharpoonup} {\mathbf{#1}} } \)

\( \newcommand{\vecd}[1]{\overset{-\!-\!\rightharpoonup}{\vphantom{a}\smash {#1}}} \)

\( \newcommand{\id}{\mathrm{id}}\) \( \newcommand{\Span}{\mathrm{span}}\)

( \newcommand{\kernel}{\mathrm{null}\,}\) \( \newcommand{\range}{\mathrm{range}\,}\)

\( \newcommand{\RealPart}{\mathrm{Re}}\) \( \newcommand{\ImaginaryPart}{\mathrm{Im}}\)

\( \newcommand{\Argument}{\mathrm{Arg}}\) \( \newcommand{\norm}[1]{\| #1 \|}\)

\( \newcommand{\inner}[2]{\langle #1, #2 \rangle}\)

\( \newcommand{\Span}{\mathrm{span}}\)

\( \newcommand{\id}{\mathrm{id}}\)

\( \newcommand{\kernel}{\mathrm{null}\,}\)

\( \newcommand{\range}{\mathrm{range}\,}\)

\( \newcommand{\RealPart}{\mathrm{Re}}\)

\( \newcommand{\ImaginaryPart}{\mathrm{Im}}\)

\( \newcommand{\Argument}{\mathrm{Arg}}\)

\( \newcommand{\norm}[1]{\| #1 \|}\)

\( \newcommand{\Span}{\mathrm{span}}\) \( \newcommand{\AA}{\unicode[.8,0]{x212B}}\)

\( \newcommand{\vectorA}[1]{\vec{#1}}      % arrow\)

\( \newcommand{\vectorAt}[1]{\vec{\text{#1}}}      % arrow\)

\( \newcommand{\vectorB}[1]{\overset { \scriptstyle \rightharpoonup} {\mathbf{#1}} } \)

\( \newcommand{\vectorC}[1]{\textbf{#1}} \)

\( \newcommand{\vectorD}[1]{\overrightarrow{#1}} \)

\( \newcommand{\vectorDt}[1]{\overrightarrow{\text{#1}}} \)

\( \newcommand{\vectE}[1]{\overset{-\!-\!\rightharpoonup}{\vphantom{a}\smash{\mathbf {#1}}}} \)

Describe the overall function of antigen-presenting cells (APCs) such as dendritic cells, macrophages, and B-lymphocytes in terms of the following: how they "process" exogenous antigens how they "process" endogenous antigens the types of MHC molecule to which they attach peptides the role of proteasomes in the binding of peptides from endogenous antigens by MHC-I molecules. the role of lysosomes in the binding of peptides from exogenous antigens by MHC-II molecules. the types of cells to which they present peptides Name the primary type of cell that functions as an antigen-presenting cell to naive T4-lymphocytes and naive T8-lymphocytes. State the role of T4-effector cells in activating macrophages. State the role of T4-effector cells in the proliferation and differentiation of activated B-lymphocytes.

alt

The primary function of dendritic cells, then, is to capture and present protein antigens to naive T-lymphocytes. (Naive lymphocytes are those that have not yet encountered an antigen.) Since dendritic cells are able to express both MHC-I and MHC-II molecules, they are able to present antigens to both naive T8-lymphocytes and naive T4-lymphocytes.

These interactions enable the naiveT4-lymphocyte or T8-lymphocyte to become activated, proliferate, and differentiate into effector lymphocytes. (Effector lymphocytes are lymphocytes that have encountered an antigen, proliferated, and matured into a form capable of actively carrying out immune defenses.)

1. MHC-II presentation of protein antigens to naive T4-lymphocytes

a. MHC-II presentation of exogenous antigens to naive T4-lymphocytes

Immature dendritic cells take in protein antigens for attachment to MHC-II molecules and subsequent presentation to naive T4-lymphocytes by:

1. Receptor-mediated phagocytosis, e.g., PAMPs binding to endocytic PRRs, IgG or C3b attachment of microbes to phagocytes during opsonization (see Figure \(\PageIndex{2}\)) .

2. Macropinocytosis, a process where large volumes of surrounding fluid containing microbes are engulfed. This also enables dendritic cells to take in some encapsulated bacteria that might resist classical phagocytosis (see Figure \(\PageIndex{3}\)) .

The binding of microbial PAMPs to the PRRs of the immature dendritic cell activates that dendritic cell and promotes production of the chemokine receptor CCR7 that directs the dendritic cell into local lymphoid tissue. Following maturation, the dendritic cell can now present protein epitopes bound to MHC molecules to all the various naive T-lymphocytes passing through the lymphoid system (See Figure \(\PageIndex{4}\) and Figure \(\PageIndex{5}\) ).

The MHC-II molecules bind peptide epitopes from exogenous antigens and place them on the surface of the dendritic cell (see Figure \(\PageIndex{6}\)) . Here the MHC-II/peptide complexes can be recognized by complementary shaped TCRs and CD4 molecules on naive T4-lymphocytes (see Figure \(\PageIndex{7}\)) .

b. MHC-II cross-presentation of endogenous antigens to naive T4-lymphocytes

While most dendritic cells present exogenous antigens to naive T4-lymphocytes, certain dendritic cells are capable of cross-presentation of endogenous antigens to naive T4-lymphocytes. In this way, T4-lymphocytes can play a role in defending against both exogenous and endogenous antigens. This is done via autophagy, the cellular process whereby the cell's own cytoplasm is taken into specialized vesicles called autophagosomes (See Figure \(\PageIndex{8}\)) . The autophagosomes subsequently fuse with lysosomes containing proteases that will degrade the proteins in the autophagosome into peptides. From here, the peptides are transported into the vesicles containing MHC-II molecules where they can bind to the MHC-II groove, be transported to the surface of the denritic cell, and interact with the TCRs and CD4 molecules of naive T4-lymphocytes (See Figure \(\PageIndex{8}\)) .

2. MHC-I presentation of protein antigens to naive T8-lymphocytes

Immature dendritic cells take in protein antigens for attachment to MHC-I molecules and subsequent presentation to naive T8-lymphocytes.

a. MHC-I presentation of endogenous antigens to naive T8-lymphocytes

During the replication of viruses and intracellular bacteria within their host cell, as well as during the replication of tumor cells, viral, bacterial, or tumor proteins are degraded into a variety of peptide epitopes by cylindrical organelles called proteasomes. The body's own cytosolic proteins are also degraded into peptides by proteasomes.

These peptide epitopes are then attached to a groove of MHC-I molecules that are then transported to the surface of that cell where they can be recognized by a complementary-shaped T-cell receptor (TCR) and a CD8 molecule, a co-receptor, on the surface of either a naive T8-lymphocyte or a cytotoxic T-lymphocyte (CTL). The TCRs recognize both the foreign peptide antigen and the MHC molecule. TCRs, however, will not recognize self-peptides bound to MHC-I. As a result, normal cells are not attacked and killed.

MHC-I molecule with bound peptide on the surface of antigen-presenting dendritic cells ; see Figure \(\PageIndex{9}\) can be recognized by a complementary-shaped TCR/CD8 on the surface of a naive T8-lymphocyte to initiate cell-mediated immunity (see Figure \(\PageIndex{10}\)) .

b. MHC-I cross-presentation of exogenous antigens to naive T8-lymphocytes

While most dendritic cells present endogenous antigens to naive T8-lymphocytes, certain dendritic cells are capable of cross-presentation of exogenous antigens to naive T8-lymphocytes. In this way, T8-lymphocytes can play a role in defending against both exogenous and endogenous antigens. There are two proposed mechanisms for cross-presentation of exogenous antigens to T8-lymphocytes:

1. The dendritic cell engulfs the exogenous antigen and places it in a phagosome which then fuses with a lysosome to form a phagolysosome. The antigen is partially degraded in the phagolysosome where proteins are translocated into the cytoplasm where they are processed into peptides by proteasomes, enter the endoplasmic reticulum, and are bound to MHC-I molecules (see Figure \(\PageIndex{11}\)) .

2. The dendritic cell engulfs the exogenous antigen and places it in a phagosome which then fuses with a lysosome to form a phagolysosome. The protein antigens are degraded into peptides within the phagolysosome which then directly fuses with vesicles containing MHC-I molecules to which the peptides subsequently bind (see Figure \(\PageIndex{12}\)) .

In addition, dendritic cells are very susceptible to infection by many different viruses. Once inside the cell, the viruses become endogenous antigens in the cytosol. Once in the cytosol, the viral proteins from the replicating viruses are degraded into peptides by proteasomes where they subsequently bind to MHC-I molecules.

The binding of microbial PAMPs to the PRRs of the immature dendritic cell activates that dendritic cell and promotes production of the chemokine receptor CCR7 that directs the dendritic cell into local lymphoid tissue. Following maturation, the dendritic cell can now present protein epitopes bound to MHC molecules to all the various naive T-lymphocytes passing through the lymphoid system.

To view an electron micrograph of a dendritic cell presenting antigen to T-lymphocytes, #1 see the Web page for the University of Illinois College of Medicine.

To view an electron micrograph of a dendritic cell presenting antigen to T-lymphocytes, #2 see the Web page for the University of Illinois College of Medicine.

Why is this essential for effective adaptive immunity ?

For a Summary of Key Surface Molecules and Cellular Interactions of Antigen-Presenting Dendritic Cells, see Figure \(\PageIndex{13}\) .

Macrophages

As we learned in Unit 5, when monocytes leave the blood and enter the tissue, they become activated and differentiate into macrophages. Those that have recently left the blood during inflammation and move to the site of infection are sometimes referred to as wandering macrophages. In addition, the body has macrophages already stationed throughout the tissues and organs of the body. These are sometimes referred to as fixed macrophages. Many fixed macrophages are part of the mononuclear phagocytic (reticuloendothelial) system. They, along with B-lymphocytes and T-lymphocytes, are found supported by reticular fibers in lymph nodules, lymph nodes, and the spleen where they filter out and phagocytose foreign matter such as microbes. Similar cells derived from stem cells, monocytes, or macrophages are also found in the liver (Kupffer cells), the kidneys (mesangial cells), the brain (microglia), the bones (osteoclasts), the lungs (alveolar macrophages), and the gastrointestinal tract (peritoneal macrophages).

The primary function of macrophages, then, is to capture and present protein antigens to effector T-lymphocytes. (Effector lymphocytes are lymphocytes that have encountered an antigen, proliferated, and matured into a form capable of actively carrying out immune defenses.)

The MHC-II molecules bind peptide epitopes from exogenous antigens and place them on the surface of the macrophages. Here the MHC-II/peptide complexes can be recognized by complementary shaped T-cell receptors (TCRs) and CD4 molecules on an effector T4-lymphocytes ; see Fig.14 .

Effector T4-lymphocytes called T H 1 cells coordinate immunity against intracellular bacteria and promote opsonization by macrophages.

1. They produce cytokines such as interferon-gamma (IFN- ? ) that promote cell-mediated immunity against intracellular pathogens, especially by activating macrophages that have either ingested pathogens or have become infected with intracellular microbes such as Mycobacterium tuberculosis, Mycobacterium leprae, Leishmania donovani , and Pneumocystis jiroveci that are able to grow in the endocytic vesicles of macrophages. Activation of the macrophage by T H 1 cells greatly enhances their antimicrobial effectiveness (see Figure \(\PageIndex{14}\)) .

2. They produce cytokines that promote the production of opsonizing and complement activating IgG that enhances phagocytosis (see Figure \(\PageIndex{15}\)) .

3. They produce receptors that bind to and kill chronically infected cells, releasing the bacteria that were growing within the cell so they can be engulfed and killed by macrophages.

4. They produce cytokines such as tumor necrosis factor-alpha (TNF-a) that promote diapedesis of macrophages.

5. They produce the chemokine CXCL2 to attract macrophages to the infection site.

There is growing evidence that monocytes and macrophages can be “trained” by an earlier infection to do better in future infections, that is, develop memory. It is thought that microbial pathogen-associated molecular patterns (PAMPs) binding to pattern-recognition (PRRs) on monocytes and macrophages triggers the cell’s epigenome to reprogram or train that cell to react better against new infections.

For a Summary of Key Surface Molecules and Cellular Interactions of Antigen-Presenting Macrophages, see Figure \(\PageIndex{16}\) .

B-lymphocytes

Like all lymphocytes, B-lymphocytes circulate back and forth between the blood and the lymphoid system of the body. B-lymphocytes are able to capture and present peptide epitopes from exogenous antigens to effector T4-lymphocytes. The MHC-II molecules bind peptide epitopes from exogenous antigens and place them on the surface of the B-lymphocytes. Here the MHC-II/peptide complexes can be recognized by complementary shaped T-cell receptors (TCRs) and CD4 molecules on an effector T4-lymphocytes (see Figure \(\PageIndex{17}\)) . This interaction eventually triggers the effector T4-lymphocyte to produce and secrete various cytokines that enable that B-lymphocyte to proliferate and differentiate into antibody-secreting plasma cells (see Figure \(\PageIndex{18}\)) .

For a Summary of Key Surface Molecules and Cellular Interactions of Antigen-Presenting B-Lymphocytes, see Figure \(\PageIndex{19}\) .

  • Antigen-presenting cells (APCs) include dendritic cells, macrophages, and B-lymphocytes.
  • APCs express both MHC-I and MHC-II molecules and serve two major functions during adaptive immunity: they capture and process antigens for presentation to T-lymphocytes, and they produce signals required for the proliferation and differentiation of lymphocytes.
  • Most dendritic cells are derived from monocytes and are referred to as myeloid dendritic cells and are located under the surface epithelium of the skin, the mucous membranes of the respiratory tract, genitourinary tract, and the gastrointestinal tract, and throughout the body's lymphoid tissues and in most solid organs.
  • The primary function of dendritic cells is to capture and present protein antigens to naive T-lymphocytes which enables the naïve T4-lymphocytes or T8-lymphocytes to become activated, proliferate, and differentiate into effector cells.
  • Naïve lymphocytes are B-lymphocytes and T-lymphocytes that have not yet reacted with an epitope of an antigen.
  • Dendritic cells use MHC-II molecules to present protein antigens to naïve T4-lymphocytes and MHC-I molecules to present protein antigens to naïve T8-lymphocytes.
  • When monocytes leave the blood and enter the tissue, they become activated and differentiate into macrophages.
  • When functioning as APCs, macrophages capture and present peptide epitopes from exogenous antigens to effector T4-lymphocytes.
  • Effector lymphocytes are lymphocytes that have encountered an antigen, proliferated, and matured into a form capable of actively carrying out immune defenses.
  • B-lymphocytes mediate antibody production.
  • When functioning as APCs, B-lymphocytes are able to capture and present peptide epitopes from exogenous antigens to effector T4-lymphocytes.
  • To activate naïve T4-lymphocytes, dendritic cells engulf exogenous antigens, place them in a phagosome, degrade protein antigens into peptides via lysosomes, bind those peptides to MHC-II molecules and transport them to the surface of the dendritic cell where they can be recognized by the T-cell receptors and CD4 molecules of naïve T4-lymphocytes.
  • To activate naïve T8-lymphocytes, dendritic cells degrade endogenous protein antigens into peptides via their proteasomes, bind those peptides to MHC-I molecules and transport them to the surface of the dendritic cell where they can be recognized by the T-cell receptors and CD8 molecules of naïve T8-lymphocytes.

Online Biology Notes

Antigen processing and presentation: Cytosolic and Endocytic pathway

August 3, 2020 Gaurab Karki Immunology 0

Antigen processing and presentation: Cytosolic and Endocytic pathway

Antigen processing and Antigen presentation

  • Antigen processing is a metabolic process that digests the proteins into peptides which can be displayed on the cell membrane together with a class-I or class-II MHC molecules and recognized by T-cells.
  • Antigen presentation is the process by which certain cell in the body especially antigen presenting cells (APCs) express processed antigen on their cell surface along with MHC molecules in the form recognizable to T cell.
  • If antigen is presented along with class-I MHC molecule, it is recognized by CD8 + Tc-cell and if presented along with class-II MHC molecule, it is recognized by CD4 + TH cells.

On the basis of types of antigen to be processed and presented, antigen processing and presenting pathway are of two types:

Cytosolic pathway of antigen processing and presentation

  • Cytosolic pathway processed and presented the endogenous antigens i.e. those generated within cell eg. Viral infected cells, tumor cells and intracellular pathogens ( M . tuberculosis, Histoplasma capsulatum).
  • The processed antigen is presented on the cell membrane with MHC-class I molecule which is recognized by CD8 + Tc-cell for degradation.

Steps involved in cytosolic pathways are:

  • Proteolytic degradation of Ag (protein) into peptides
  • Transportation of peptides from cytosol to RER
  • Assembly of peptides with class I MHC molecules

i. Proteolytic degradation of proteins into peptides:

  • Intracellular proteineous antigen are larger in size to be bound to MHC molecule.
  • So, it is degraded into short peptides of about 8-10 amino acids.
  • These proteins are degraded by cytosolic proteolytic system present in cell called proteasome.
  • The large (20S) proteasome is composed of 14 sub-units arranged in barrel-like structure of symmetrical rings.
  • Some, but not all the sub-units have protease activity.
  • Proteins enter the proteasome through narrow channel at each end.
  • Many proteins targeted for proteolysis have a small protein called ubiquitin attached to them.
  • Ubiquitin attached to them ubiquitin-protein complex consisting of 20S proteasome and 19S regulatory component added to it.
  • The resulting 26S proteasome cleaves peptide bonds which is ATP-dependent process.
  • Degradation of ubiquitin protein complex is thought to occur within the central hollow of the proteasome to release peptides.

ii. Transportation of peptides from cytosol to Rough Endoplasmic Reticulum (RER):

  • Peptides generated in cytosol by proteasome are transported by TAP (transporter associated with antigen processing) into RER (Rough endoplasmic reticulum) by a process which require hydrolysis of ATP.
  • TAP is membrane spanning heterodimer consisting of two proteins, TAP1 and TAP2.
  • TAP has affinity for peptides having 8-16 amino acids.
  • The optimal peptide length required by class-I MHC for binding is nine, which is achieved by trimming the peptides with the help of amino-peptidase present in RER. Eg. ERAP.
  • In addition to it, TAP favor peptides with hydrophobic or basic carboxyl terminal amino acids, that preferred anchor residues for class-I MHC molecules.
  • TAP deficiency can lead to a disease syndrome that has both immune-deficiency and auto-immunity aspects.

iii. Assembly of peptides with class-I MHC molecule:

  • Like other proteins, the α-chain and β 2 microglobulin components of the class-I MHC molecule are synthesized on polysome along the rough endoplasmic reticulum.
  • Assembly of these components into stable class-I MHC molecule that can exit the RRE require binding of peptides into peptide binding groove of class-I MHC molecules.
  • The assembly process involves several steps and needs help of molecular chaperone.
  • The first molecular chaperone involved in assembly of class-I MHC is calnexin.
  • It is a resident membrane protein of RER.
  • Calnexin associated with free class-I α-chain and promotes its folding.
  • When β 2 -microglobulin binds class-I α-chain, calnexin is released and class-I MHC associates with another chaperone calreticulin and tapasin (TAP-associated protein).
  • Tapasin brings TAP transporter carrying peptides to the proximity with class-I MHC molecule and allows to acquire the antigenic peptides.
  • An additional protein with enzymatic activity, ERp57, form disulfide bond to tapasin and non-covalently associates with calreticulin to stabilize the interaction and allows release of MHC-I-class after acquiring antigenic peptides.
  • As a consequence, the productive peptide binding with MHC of class-I releases from the complex of calreticulin, tapasin and ERp57, exit from RER and displays on the cell surface via golgi complex.

explain the process of antigen presentation

Endocytic pathway of antigen processing and presentation:

  • The endocytic pathway processed and present the exogenous Ag. i.e. antigens generated outside the cells. E.g. Bacteria.
  • At first APC phagocytosed, endocytosed or both, the antigen.
  • Macrophage and dendritic cells internalize the antigen by both the process.
  • While other APCs are non-phagocytic or poorly phagocytic. E.g. B cell internalize the antigen by receptor mediated endocytosis.
  • Then antigen is processed and presented on the cell surface along with class-II MHC molecules which are recognized by CD4 + TH cell.

Steps involved in endocytic pathway:

  • Peptide generation from internalized molecules (Ag) in endocytic vesicles.
  • Transport of class-II MHC molecule to endocytic vesicles.
  • Assembly of peptides with Class-II MHC molecules.

i. Peptide generation from internalized molecules (Ag) in endocytic vesicles:

  • Once an antigen is internalized, it is degraded into peptides within compartments of endocytic processing pathway.
  • The endocytic pathway appears to involve three increasingly acidic compartments, early endosomes (pH 6-6.5), late endosomes or endo-lysosome (pH 5-6) and lysosomes (pH 4.5-5).
  • The internalized antigens move from early to late endosomes and finally to lysosomes, encountering hydrolytic enzymes and a lower pH in each compartment.
  • Within the compartment, antigen is degraded into oligopeptides of about 13-18 residues.
  • The mechanism by which internalized Ag moves from one endocytic compartment to next has not been clearly demonstrated.
  • It has been suggested that early endosome move from periphery to inward to become late endosome and finally lysosomes.
  • Alternatively, small transport vesicles may carry Ag from one compartment to next.

ii. Transport of class-II MHC molecule to endocytic vesicles:

  • When class-II MHC molecules are synthesized within RER, three pairs of class-II αβ- chains associated with a pre-assembled trimer of a protein called invariant chain (Li, CD74).
  • This trimeric protein prevents any endogenously antigen to bind to the cleft.
  • The invariant chain consists of sorting signals in its cytoplasmic tail.
  • It directs the transport of class-II MHC molecule to endocytic compartments from the trans-golgi network.

iii. Assembly of peptides with class-II MHC molecules:

  • Class-II MHC-invariant chain complexes are transported from RER through golgi complex and golgi-network and through endocytic compartment, moving from early endosome to late endosome and finally to lysosome.
  • The proteolytic activities increase in each compartment, so the invariant is slowly degraded.
  • However, a short fragment of invariant chain remained termed as CLIP (Class-II associated invariant chain).
  • CLIP physically occupies the peptide binding, cleft of class-II MHC molecule, presumably preventing any premature binding of antigenic peptides.
  • A non-classical class-II MHC molecule known as HLA-DM is required to catalyze the exchange of CLIP with antigenic peptides.
  • The reaction between HLA-DO, which binds to HLA-DM and lessens the efficiency of the exchange reactions.
  • Conditions of higher acidity in endocytic compartment weakens the association of DM/DO and increase the possibility of antigenic peptide binding despite of DO.
  • As with class-I MHC molecule, peptide binding is required to maintain the structure and stability of class-II MHC molecules.
  • Once a peptide has bound the peptide-class II MHC complex is transported to the plasma membrane where neutral pH enables the complex to assume the compact and stable form.

explain the process of antigen presentation

  • Antigen processing and presentationCytosolic and Endocytic pathway

Copyright © 2024 | WordPress Theme by MH Themes

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Review Article
  • Published: 16 June 2023

Antigen presentation in cancer — mechanisms and clinical implications for immunotherapy

  • Kailin Yang 1 ,
  • Ahmed Halima 1 &
  • Timothy A. Chan   ORCID: orcid.org/0000-0002-9265-0283 1 , 2 , 3 , 4  

Nature Reviews Clinical Oncology volume  20 ,  pages 604–623 ( 2023 ) Cite this article

15k Accesses

22 Citations

28 Altmetric

Metrics details

  • Immunotherapy
  • Predictive markers
  • Tumour immunology

Over the past decade, the emergence of effective immunotherapies has revolutionized the clinical management of many types of cancers. However, long-term durable tumour control is only achieved in a fraction of patients who receive these therapies. Understanding the mechanisms underlying clinical response and resistance to treatment is therefore essential to expanding the level of clinical benefit obtained from immunotherapies. In this Review, we describe the molecular mechanisms of antigen processing and presentation in tumours and their clinical consequences. We examine how various aspects of the antigen-presentation machinery (APM) shape tumour immunity. In particular, we discuss genomic variants in HLA alleles and other APM components, highlighting their influence on the immunopeptidomes of both malignant cells and immune cells. Understanding the APM, how it is regulated and how it changes in tumour cells is crucial for determining which patients will respond to immunotherapy and why some patients develop resistance. We focus on recently discovered molecular and genomic alterations that drive the clinical outcomes of patients receiving immune-checkpoint inhibitors. An improved understanding of how these variables mediate tumour–immune interactions is expected to guide the more precise administration of immunotherapies and reveal potentially promising directions for the development of new immunotherapeutic approaches.

The clinical success of immune-checkpoint inhibitors has improved cancer care, although long-term durable remission is only achieved in a subset of patients.

Antigen processing and presentation by tumour cells are essential for long-lasting immune surveillance.

Alterations in the genes encoding MHC components and other parts of the antigen-presentation machinery are frequently found across several cancer types and are associated with both tumour development and the effectiveness of immunotherapies.

MHC-based antigen presentation exerts strong evolutionary pressure on the immunopeptidome, which in turn shapes the mutational landscape of the tumour genome.

Germline human leukocyte antigen diversity and somatic aberrations in the antigen-presentation machinery inform the therapeutic response to immune-checkpoint inhibitors.

Development of novel therapies based on an accurate understanding of antigen presentation in the setting of tumour–immune dynamics is crucial to the development of improved therapeutic approaches.

This is a preview of subscription content, access via your institution

Access options

Access Nature and 54 other Nature Portfolio journals

Get Nature+, our best-value online-access subscription

24,99 € / 30 days

cancel any time

Subscribe to this journal

Receive 12 print issues and online access

195,33 € per year

only 16,28 € per issue

Buy this article

  • Purchase on Springer Link
  • Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

explain the process of antigen presentation

Similar content being viewed by others

explain the process of antigen presentation

Antigen presentation in cancer: insights into tumour immunogenicity and immune evasion

explain the process of antigen presentation

Towards new horizons: characterization, classification and implications of the tumour antigenic repertoire

explain the process of antigen presentation

Therapeutic cancer vaccines: advancements, challenges, and prospects

Waldman, A. D., Fritz, J. M. & Lenardo, M. J. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat. Rev. Immunol. 20 , 651–668 (2020).

Article   CAS   PubMed   PubMed Central   Google Scholar  

Hodi, F. S. et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 363 , 711–723 (2010).

Larkin, J. et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 373 , 23–34 (2015).

Article   PubMed   PubMed Central   Google Scholar  

Schadendorf, D. et al. Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma. J. Clin. Oncol. 33 , 1889–1894 (2015).

Robert, C. et al. Nivolumab in previously untreated melanoma without BRAF mutation. N. Engl. J. Med. 372 , 320–330 (2015).

Article   CAS   PubMed   Google Scholar  

Borghaei, H. et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N. Engl. J. Med. 373 , 1627–1639 (2015).

Motzer, R. J. et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N. Engl. J. Med. 373 , 1803–1813 (2015).

Burtness, B. et al. Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): a randomised, open-label, phase 3 study. Lancet 394 , 1915–1928 (2019).

Bellmunt, J. et al. Adjuvant atezolizumab versus observation in muscle-invasive urothelial carcinoma (IMvigor010): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 22 , 525–537 (2021).

Kelly, R. J. et al. Adjuvant nivolumab in resected esophageal or gastroesophageal junction cancer. N. Engl. J. Med. 384 , 1191–1203 (2021).

Schmid, P. et al. Atezolizumab and Nab-paclitaxel in advanced triple-negative breast cancer. N. Engl. J. Med. 379 , 2108–2121 (2018).

Ribas, A. & Wolchok, J. D. Cancer immunotherapy using checkpoint blockade. Science 359 , 1350–1355 (2018).

Antonia, S. J. et al. Overall survival with durvalumab after chemoradiotherapy in stage III NSCLC. N. Engl. J. Med. 379 , 2342–2350 (2018).

Forde, P. M. et al. Neoadjuvant nivolumab plus chemotherapy in resectable lung cancer. N. Engl. J. Med. 386 , 1973–1985 (2022).

Snyder, A. et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 371 , 2189–2199 (2014).

Gao, J. et al. Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell 167 , 397–404.e9 (2016).

Zaretsky, J. M. et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N. Engl. J. Med. 375 , 819–829 (2016).

Zhou, X. et al. Treatment-related adverse events of PD-1 and PD-L1 inhibitor-based combination therapies in clinical trials: a systematic review and meta-analysis. Lancet Oncol. 22 , 1265–1274 (2021).

Havel, J. J., Chowell, D. & Chan, T. A. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat. Rev. Cancer 19 , 133–150 (2019).

Halima, A., Vuong, W. & Chan, T. A. Next-generation sequencing: unraveling genetic mechanisms that shape cancer immunotherapy efficacy. J. Clin. Invest. 132 , e154945 (2022).

Pietanza, M. C. et al. Phase II trial of temozolomide in patients with relapsed sensitive or refractory small cell lung cancer, with assessment of methylguanine-DNA methyltransferase as a potential biomarker. Clin. Cancer Res. 18 , 1138–1145 (2012).

Rizvi, N. A. et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348 , 124–128 (2015).

Dudley, J. C., Lin, M. T., Le, D. T. & Eshleman, J. R. Microsatellite instability as a biomarker for PD-1 blockade. Clin. Cancer Res. 22 , 813–820 (2016).

Le, D. T. et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357 , 409–413 (2017).

Marcus, L., Lemery, S. J., Keegan, P. & Pazdur, R. FDA approval summary: pembrolizumab for the treatment of microsatellite instability-high solid tumors. Clin. Cancer Res. 25 , 3753–3758 (2019).

Cercek, A. et al. PD-1 blockade in mismatch repair-deficient, locally advanced rectal cancer. N. Engl. J. Med. 386 , 2363–2376 (2022).

Rosenberg, J. E. et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet 387 , 1909–1920 (2016).

Gong, J., Chehrazi-Raffle, A., Reddi, S. & Salgia, R. Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. J. Immunother. Cancer 6 , 8 (2018).

Chan, T. A. et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann. Oncol. 30 , 44–56 (2019).

Mahanty, S., Prigent, A. & Garraud, O. Immunogenicity of infectious pathogens and vaccine antigens. BMC Immunol. 16 , 31 (2015).

Pollard, A. J. & Bijker, E. M. A guide to vaccinology: from basic principles to new developments. Nat. Rev. Immunol. 21 , 83–100 (2021).

Li, F. et al. The association between CD8 + tumor-infiltrating lymphocytes and the clinical outcome of cancer immunotherapy: a systematic review and meta-analysis. EClinicalMedicine 41 , 101134 (2021).

Hellstrom, I., Hellstrom, K. E., Pierce, G. E. & Yang, J. P. Cellular and humoral immunity to different types of human neoplasms. Nature 220 , 1352–1354 (1968).

Horton, B. L. et al. Lack of CD8 + T cell effector differentiation during priming mediates checkpoint blockade resistance in non-small cell lung cancer. Sci. Immunol. 6 , eabi8800 (2021).

Lee, M. Y., Jeon, J. W., Sievers, C. & Allen, C. T. Antigen processing and presentation in cancer immunotherapy. J. Immunother. Cancer 8 , e001111 (2020).

Doherty, P. C. & Zinkernagel, R. M. A biological role for the major histocompatibility antigens. Lancet 1 , 1406–1409 (1975).

Doherty, P. C. & Zinkernagel, R. M. Enhanced immunological surveillance in mice heterozygous at the H-2 gene complex. Nature 256 , 50–52 (1975).

Parham, P. & Ohta, T. Population biology of antigen presentation by MHC class I molecules. Science 272 , 67–74 (1996).

Matsumura, M., Fremont, D. H., Peterson, P. A. & Wilson, I. A. Emerging principles for the recognition of peptide antigens by MHC class I molecules. Science 257 , 927–934 (1992).

Paul, S. et al. HLA class I alleles are associated with peptide-binding repertoires of different size, affinity, and immunogenicity. J. Immunol. 191 , 5831–5839 (2013).

Neefjes, J., Jongsma, M. L., Paul, P. & Bakke, O. Towards a systems understanding of MHC class I and MHC class II antigen presentation. Nat. Rev. Immunol. 11 , 823–836 (2011).

Colbert, J. D., Cruz, F. M. & Rock, K. L. Cross-presentation of exogenous antigens on MHC I molecules. Curr. Opin. Immunol. 64 , 1–8 (2020).

Chicz, R. M. et al. Predominant naturally processed peptides bound to HLA-DR1 are derived from MHC-related molecules and are heterogeneous in size. Nature 358 , 764–768 (1992).

Roche, P. A. & Furuta, K. The ins and outs of MHC class II-mediated antigen processing and presentation. Nat. Rev. Immunol. 15 , 203–216 (2015).

Dendrou, C. A., Petersen, J., Rossjohn, J. & Fugger, L. HLA variation and disease. Nat. Rev. Immunol. 18 , 325–339 (2018).

Arnaiz-Villena, A. et al. Evolution and molecular interactions of major histocompatibility complex (MHC)-G, -E and -F genes. Cell Mol. Life Sci. 79 , 464 (2022).

Baumeister, W., Walz, J., Zuhl, F. & Seemuller, E. The proteasome: paradigm of a self-compartmentalizing protease. Cell 92 , 367–380 (1998).

Groll, M. et al. Structure of 20S proteasome from yeast at 2.4 A resolution. Nature 386 , 463–471 (1997).

Dong, Y. et al. Cryo-EM structures and dynamics of substrate-engaged human 26S proteasome. Nature 565 , 49–55 (2019).

Cascio, P., Hilton, C., Kisselev, A. F., Rock, K. L. & Goldberg, A. L. 26S proteasomes and immunoproteasomes produce mainly N-extended versions of an antigenic peptide. EMBO J. 20 , 2357–2366 (2001).

Vigneron, N. et al. An antigenic peptide produced by peptide splicing in the proteasome. Science 304 , 587–590 (2004).

Warren, E. H. et al. An antigen produced by splicing of noncontiguous peptides in the reverse order. Science 313 , 1444–1447 (2006).

Rolfs, Z., Muller, M., Shortreed, M. R., Smith, L. M. & Bassani-Sternberg, M. Comment on “A subset of HLA-I peptides are not genomically templated: evidence for cis- and trans-spliced peptide ligands”. Sci. Immunol. 4 , eaaw1622 (2019).

Admon, A. Are there indeed spliced peptides in the immunopeptidome? Mol. Cell Proteom. 20 , 100099 (2021).

Article   CAS   Google Scholar  

Rock, K. L. & Goldberg, A. L. Degradation of cell proteins and the generation of MHC class I-presented peptides. Annu. Rev. Immunol. 17 , 739–779 (1999).

Vigneron, N. & Van den Eynde, B. J. Proteasome subtypes and the processing of tumor antigens: increasing antigenic diversity. Curr. Opin. Immunol. 24 , 84–91 (2012).

Parcej, D. & Tampe, R. ABC proteins in antigen translocation and viral inhibition. Nat. Chem. Biol. 6 , 572–580 (2010).

Serwold, T., Gonzalez, F., Kim, J., Jacob, R. & Shastri, N. ERAAP customizes peptides for MHC class I molecules in the endoplasmic reticulum. Nature 419 , 480–483 (2002).

Pishesha, N., Harmand, T. J. & Ploegh, H. L. A guide to antigen processing and presentation. Nat. Rev. Immunol. https://doi.org/10.1038/s41577-022-00707-2 (2022).

Article   PubMed   Google Scholar  

Garstka, M. A. et al. The first step of peptide selection in antigen presentation by MHC class I molecules. Proc. Natl Acad. Sci. USA 112 , 1505–1510 (2015).

Zarling, A. L. et al. Tapasin is a facilitator, not an editor, of class I MHC peptide binding. J. Immunol. 171 , 5287–5295 (2003).

Hermann, C. et al. TAPBPR alters MHC class I peptide presentation by functioning as a peptide exchange catalyst. eLife 4 , e09617 (2015).

Hildner, K. et al. Batf3 deficiency reveals a critical role for CD8α + dendritic cells in cytotoxic T cell immunity. Science 322 , 1097–1100 (2008).

Bonaccorsi, I. et al. Membrane transfer from tumor cells overcomes deficient phagocytic ability of plasmacytoid dendritic cells for the acquisition and presentation of tumor antigens. J. Immunol. 192 , 824–832 (2014).

Sanchez-Paulete, A. R. et al. Antigen cross-presentation and T-cell cross-priming in cancer immunology and immunotherapy. Ann. Oncol. 28 , xii44–xii55 (2017).

Blander, J. M. Regulation of the cell biology of antigen cross-presentation. Annu. Rev. Immunol. 36 , 717–753 (2018).

Canton, J. et al. The receptor DNGR-1 signals for phagosomal rupture to promote cross-presentation of dead-cell-associated antigens. Nat. Immunol. 22 , 140–153 (2021).

Shen, L., Sigal, L. J., Boes, M. & Rock, K. L. Important role of cathepsin S in generating peptides for TAP-independent MHC class I crosspresentation in vivo. Immunity 21 , 155–165 (2004).

Sanchez-Paulete, A. R. et al. Cancer immunotherapy with immunomodulatory anti-CD137 and anti-PD-1 monoclonal antibodies requires BATF3-dependent dendritic cells. Cancer Discov. 6 , 71–79 (2016).

Hanahan, D. Hallmarks of cancer: new dimensions. Cancer Discov. 12 , 31–46 (2022).

Lipsitch, M., Bergstrom, C. T. & Antia, R. Effect of human leukocyte antigen heterozygosity on infectious disease outcome: the need for allele-specific measures. BMC Med. Genet. 4 , 2 (2003).

Hraber, P., Kuiken, C. & Yusim, K. Evidence for human leukocyte antigen heterozygote advantage against hepatitis C virus infection. Hepatology 46 , 1713–1721 (2007).

Arora, J. et al. HLA heterozygote advantage against HIV-1 is driven by quantitative and qualitative differences in HLA allele-specific peptide presentation. Mol. Biol. Evol. 37 , 639–650 (2020).

Liu, Z. et al. HLA zygosity increases risk of hepatitis B virus-associated hepatocellular carcinoma. J. Infect. Dis. 224 , 1796–1805 (2021).

Gubin, M. M. et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 515 , 577–581 (2014).

Tran, E. et al. Immunogenicity of somatic mutations in human gastrointestinal cancers. Science 350 , 1387–1390 (2015).

Chowell, D. et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 359 , 582–587 (2018).

Anagnostou, V. et al. Multimodal genomic features predict outcome of immune checkpoint blockade in non-small-cell lung cancer. Nat. Cancer 1 , 99–111 (2020).

Abed, A. et al. Prognostic value of HLA-I homozygosity in patients with non-small cell lung cancer treated with single agent immunotherapy. J. Immunother. Cancer 8 , e001620 (2020).

Kobayashi, M. et al. Effect of HLA genotype on intravesical recurrence after bacillus Calmette-Guerin therapy for non-muscle-invasive bladder cancer. Cancer Immunol. Immunother. 71 , 727–736 (2022).

Chowell, D. et al. Evolutionary divergence of HLA class I genotype impacts efficacy of cancer immunotherapy. Nat. Med. 25 , 1715–1720 (2019).

Chowell, D. et al. Improved prediction of immune checkpoint blockade efficacy across multiple cancer types. Nat. Biotechnol. 40 , 499–506 (2022).

Litchfield, K. et al. Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition. Cell 184 , 596–614.e14 (2021).

Cuppens, K. et al. HLA-I diversity and tumor mutational burden by comprehensive next-generation sequencing as predictive biomarkers for the treatment of non-small cell lung cancer with PD-(L)1 inhibitors. Lung Cancer 170 , 1–10 (2022).

Lu, Z. et al. Germline HLA-B evolutionary divergence influences the efficacy of immune checkpoint blockade therapy in gastrointestinal cancer. Genome Med. 13 , 175 (2021).

Lee, C. H. et al. High response rate and durability driven by HLA genetic diversity in patients with kidney cancer treated with Lenvatinib and Pembrolizumab. Mol. Cancer Res. 19 , 1510–1521 (2021).

Takahashi, S. et al. Impact of germline HLA genotypes on clinical outcomes in patients with urothelial cancer treated with pembrolizumab. Cancer Sci. https://doi.org/10.1111/cas.15488 (2022).

Roerden, M. et al. HLA evolutionary divergence as a prognostic marker for AML patients undergoing allogeneic stem cell transplantation. Cancers 12 , 1835 (2020).

Feray, C. et al. Donor HLA class 1 evolutionary divergence is a major predictor of liver allograft rejection: a retrospective cohort study. Ann. Intern. Med. 174 , 1385–1394 (2021).

Cummings, A. L. et al. Mutational landscape influences immunotherapy outcomes among patients with non-small-cell lung cancer with human leukocyte antigen supertype B44. Nat. Cancer 1 , 1167–1175 (2020).

Naranbhai, V. et al. HLA-A*03 and response to immune checkpoint blockade in cancer: an epidemiological biomarker study. Lancet Oncol. 23 , 172–184 (2022).

Pyke, R. M. et al. A machine learning algorithm with subclonal sensitivity reveals widespread pan-cancer human leukocyte antigen loss of heterozygosity. Nat. Commun. 13 , 1925 (2022).

Hazini, A., Fisher, K. & Seymour, L. Deregulation of HLA-I in cancer and its central importance for immunotherapy. J. Immunother. Cancer 9 , e002899 (2021).

Shukla, S. A. et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat. Biotechnol. 33 , 1152–1158 (2015).

Salter, R. D. et al. A binding site for the T-cell co-receptor CD8 on the α 3 domain of HLA-A2. Nature 345 , 41–46 (1990).

Alspach, E. et al. MHC-II neoantigens shape tumour immunity and response to immunotherapy. Nature 574 , 696–701 (2019).

Shao, X. M. et al. HLA class II immunogenic mutation burden predicts response to immune checkpoint blockade. Ann. Oncol. 33 , 728–738 (2022).

Pagliuca, S. et al. The similarity of class II HLA genotypes defines patterns of autoreactivity in idiopathic bone marrow failure disorders. Blood 138 , 2781–2798 (2021).

Daull, A. M. et al. Class I/Class II HLA evolutionary divergence ratio is an independent marker associated with disease-free and overall survival after allogeneic hematopoietic stem cell transplantation for acute myeloid leukemia. Front. Immunol. 13 , 841470 (2022).

Steimle, V., Siegrist, C. A., Mottet, A., Lisowska-Grospierre, B. & Mach, B. Regulation of MHC class II expression by interferon-γ mediated by the transactivator gene CIITA. Science 265 , 106–109 (1994).

Rodig, S. J. et al. MHC proteins confer differential sensitivity to CTLA-4 and PD-1 blockade in untreated metastatic melanoma. Sci. Transl. Med. 10 , eaar3342 (2018).

Michelakos, T. et al. Differential role of HLA-A and HLA-B, C expression levels as prognostic markers in colon and rectal cancer. J. Immunother. Cancer 10 , e004115 (2022).

Meissner, T. B. et al. NLR family member NLRC5 is a transcriptional regulator of MHC class I genes. Proc. Natl Acad. Sci. USA 107 , 13794–13799 (2010).

Yoshihama, S. et al. NLRC5/MHC class I transactivator is a target for immune evasion in cancer. Proc. Natl Acad. Sci. USA 113 , 5999–6004 (2016).

Chew, G. L. et al. DUX4 suppresses MHC class I to promote cancer immune evasion and resistance to checkpoint blockade. Dev. Cell 50 , 658–671.e7 (2019).

Forloni, M. et al. NF-κB, and not MYCN, regulates MHC class I and endoplasmic reticulum aminopeptidases in human neuroblastoma cells. Cancer Res. 70 , 916–924 (2010).

Lorenzi, S. et al. IRF1 and NF-kB restore MHC class I-restricted tumor antigen processing and presentation to cytotoxic T cells in aggressive neuroblastoma. PLoS ONE   7 , e46928 (2012).

Yi, M. et al. The role of cancer-derived microRNAs in cancer immune escape. J. Hematol. Oncol. 13 , 25 (2020).

Mari, L. et al. microRNA 125a regulates MHC-I expression on esophageal adenocarcinoma cells, associated with suppression of antitumor immune response and poor outcomes of patients. Gastroenterology 155 , 784–798 (2018).

Colangelo, T. et al. Proteomic screening identifies calreticulin as a miR-27a direct target repressing MHC class I cell surface exposure in colorectal cancer. Cell Death Dis. 7 , e2120 (2016).

Huang, L. et al. The RNA-binding protein MEX3B mediates resistance to cancer immunotherapy by downregulating HLA-A expression. Clin. Cancer Res. 24 , 3366–3376 (2018).

Cano, F., Rapiteanu, R., Winkler, G. S. & Lehner, P. J. A non-proteolytic role for ubiquitin in deadenylation of MHC-I mRNA by the RNA-binding E3-ligase MEX-3C. Nat. Commun. 6 , 8670 (2015).

Yamamoto, K. et al. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Nature 581 , 100–105 (2020).

Wang, Y. et al. Oncoprotein SND1 hijacks nascent MHC-I heavy chain to ER-associated degradation, leading to impaired CD8 + T cell response in tumor. Sci. Adv. 6 , eaba5412 (2020).

Fang, Y. et al. MAL2 drives immune evasion in breast cancer by suppressing tumor antigen presentation. J. Clin. Invest. 131 , e140837 (2021).

Jongsma, M. L. M. et al. The SPPL3-defined glycosphingolipid repertoire orchestrates HLA class I-mediated immune responses. Immunity 54 , 132–150.e9 (2021).

Rooney, M. S., Shukla, S. A., Wu, C. J., Getz, G. & Hacohen, N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell 160 , 48–61 (2015).

Bernal, M., Ruiz-Cabello, F., Concha, A., Paschen, A. & Garrido, F. Implication of the β2-microglobulin gene in the generation of tumor escape phenotypes. Cancer Immunol. Immunother. 61 , 1359–1371 (2012).

Gettinger, S. et al. Impaired HLA class I antigen processing and presentation as a mechanism of acquired resistance to immune checkpoint inhibitors in lung cancer. Cancer Discov. 7 , 1420–1435 (2017).

Sade-Feldman, M. et al. Resistance to checkpoint blockade therapy through inactivation of antigen presentation. Nat. Commun. 8 , 1136 (2017).

Challa-Malladi, M. et al. Combined genetic inactivation of β2-microglobulin and CD58 reveals frequent escape from immune recognition in diffuse large B cell lymphoma. Cancer Cell 20 , 728–740 (2011).

Fangazio, M. et al. Genetic mechanisms of HLA-I loss and immune escape in diffuse large B cell lymphoma. Proc. Natl Acad. Sci. USA 118 , e2104504118 (2021).

Chen, H. L. et al. A functionally defective allele of TAP1 results in loss of MHC class I antigen presentation in a human lung cancer. Nat. Genet. 13 , 210–213 (1996).

Klampfl, T. et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N. Engl. J. Med. 369 , 2379–2390 (2013).

Arshad, N. & Cresswell, P. Tumor-associated calreticulin variants functionally compromise the peptide loading complex and impair its recruitment of MHC-I. J. Biol. Chem. 293 , 9555–9569 (2018).

Tripathi, S. C. et al. Immunoproteasome deficiency is a feature of non-small cell lung cancer with a mesenchymal phenotype and is associated with a poor outcome. Proc. Natl Acad. Sci. USA 113 , E1555–E1564 (2016).

Kalaora, S. et al. Immunoproteasome expression is associated with better prognosis and response to checkpoint therapies in melanoma. Nat. Commun. 11 , 896 (2020).

Ramsuran, V. et al. Epigenetic regulation of differential HLA-A allelic expression levels. Hum. Mol. Genet. 24 , 4268–4275 (2015).

Ye, Q. et al. Hypermethylation of HLA class I gene is associated with HLA class I down-regulation in human gastric cancer. Tissue Antigens 75 , 30–39 (2010).

Burr, M. L. et al. An evolutionarily conserved function of polycomb silences the MHC class I antigen presentation pathway and enables immune evasion in cancer. Cancer Cell 36 , 385–401.e8 (2019).

Ennishi, D. et al. Molecular and genetic characterization of MHC deficiency identifies EZH2 as therapeutic target for enhancing immune recognition. Cancer Discov. 9 , 546–563 (2019).

Guo, B., Tan, X. & Cen, H. EZH2 is a negative prognostic biomarker associated with immunosuppression in hepatocellular carcinoma. PLoS ONE   15 , e0242191 (2020).

He, P. C. & He, C. m 6 A RNA methylation: from mechanisms to therapeutic potential. EMBO J. 40 , e105977 (2021).

McFadden, M. J. et al. Post-transcriptional regulation of antiviral gene expression by N6-methyladenosine. Cell Rep. 34 , 108798 (2021).

Jin, S. et al. m 6 A RNA modification controls autophagy through upregulating ULK1 protein abundance. Cell Res. 28 , 955–957 (2018).

Wang, X. et al. m 6 A mRNA methylation controls autophagy and adipogenesis by targeting Atg5 and Atg7. Autophagy 16 , 1221–1235 (2020).

Han, D. et al. Anti-tumour immunity controlled through mRNA m 6 A methylation and YTHDF1 in dendritic cells. Nature 566 , 270–274 (2019).

Vizcaino, J. A. et al. The human immunopeptidome project: a roadmap to predict and treat immune diseases. Mol. Cell Proteom. 19 , 31–49 (2020).

Marcu, A. et al. HLA ligand atlas: a benign reference of HLA-presented peptides to improve T-cell-based cancer immunotherapy. J. Immunother. Cancer 9 , e002071 (2021).

Kubiniok, P. et al. Understanding the constitutive presentation of MHC class I immunopeptidomes in primary tissues. iScience 25 , 103768 (2022).

Phillips, E. R. & Perdue, J. F. The expression and localization of surface neoantigens in transformed and untransformed cultured cells infected with avian tumor viruses. J. Supramol. Struct. 4 , 27–44 (1976).

Lennerz, V. et al. The response of autologous T cells to a human melanoma is dominated by mutated neoantigens. Proc. Natl Acad. Sci. USA 102 , 16013–16018 (2005).

Maletzki, C., Schmidt, F., Dirks, W. G., Schmitt, M. & Linnebacher, M. Frameshift-derived neoantigens constitute immunotherapeutic targets for patients with microsatellite-instable haematological malignancies: frameshift peptides for treating MSI + blood cancers. Eur. J. Cancer 49 , 2587–2595 (2013).

McGranahan, N. et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 351 , 1463–1469 (2016).

Yang, W. et al. Immunogenic neoantigens derived from gene fusions stimulate T cell responses. Nat. Med. 25 , 767–775 (2019).

Khodadoust, M. S. et al. Antigen presentation profiling reveals recognition of lymphoma immunoglobulin neoantigens. Nature 543 , 723–727 (2017).

Haen, S. P., Loffler, M. W., Rammensee, H. G. & Brossart, P. Towards new horizons: characterization, classification and implications of the tumour antigenic repertoire. Nat. Rev. Clin. Oncol. 17 , 595–610 (2020).

Kreiter, S. et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 520 , 692–696 (2015).

Kalaora, S. et al. Use of HLA peptidomics and whole exome sequencing to identify human immunogenic neo-antigens. Oncotarget 7 , 5110–5117 (2016).

Ott, P. A. et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 547 , 217–221 (2017).

Keskin, D. B. et al. Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature 565 , 234–239 (2019).

Platten, M. et al. A vaccine targeting mutant IDH1 in newly diagnosed glioma. Nature 592 , 463–468 (2021).

Palmer, C. D. et al. Individualized, heterologous chimpanzee adenovirus and self-amplifying mRNA neoantigen vaccine for advanced metastatic solid tumors: phase 1 trial interim results. Nat. Med. 28 , 1619–1629 (2022).

Rotzschke, O. et al. Isolation and analysis of naturally processed viral peptides as recognized by cytotoxic T cells. Nature 348 , 252–254 (1990).

Hunt, D. F. et al. Characterization of peptides bound to the class I MHC molecule HLA-A2.1 by mass spectrometry. Science 255 , 1261–1263 (1992).

Bassani-Sternberg, M. et al. Direct identification of clinically relevant neoepitopes presented on native human melanoma tissue by mass spectrometry. Nat. Commun. 7 , 13404 (2016).

Schuster, H. et al. The immunopeptidomic landscape of ovarian carcinomas. Proc. Natl Acad. Sci. USA 114 , E9942–E9951 (2017).

Shao, W. et al. The SysteMHC Atlas project. Nucleic Acids Res. 46 , D1237–D1247 (2018).

Yi, X. et al. caAtlas: an immunopeptidome atlas of human cancer. iScience 24 , 103107 (2021).

Stopfer, L. E. et al. Absolute quantification of tumor antigens using embedded MHC-I isotopologue calibrants. Proc. Natl Acad. Sci. USA 118 , e2111173118 (2021).

Jaeger, A. M. et al. Deciphering the immunopeptidome in vivo reveals new tumour antigens. Nature 607 , 149–155 (2022).

Kalaora, S. et al. Identification of bacteria-derived HLA-bound peptides in melanoma. Nature 592 , 138–143 (2021).

Nielsen, M., Ternette, N. & Barra, C. The interdependence of machine learning and LC-MS approaches for an unbiased understanding of the cellular immunopeptidome. Expert. Rev. Proteom. 19 , 77–88 (2022).

Bassani-Sternberg, M. et al. Deciphering HLA-I motifs across HLA peptidomes improves neo-antigen predictions and identifies allostery regulating HLA specificity. PLoS Comput. Biol. 13 , e1005725 (2017).

Andreatta, M. et al. MS-Rescue: a computational pipeline to increase the quality and yield of immunopeptidomics experiments. Proteomics 19 , e1800357 (2019).

Alvarez, B. et al. NNAlign_MA; MHC peptidome deconvolution for accurate MHC binding motif characterization and improved T-cell epitope predictions. Mol. Cell Proteom. 18 , 2459–2477 (2019).

Jurtz, V. et al. NetMHCpan-4.0: improved peptide-MHC class I interaction predictions integrating eluted ligand and peptide binding affinity data. J. Immunol. 199 , 3360–3368 (2017).

O’Donnell, T. J., Rubinsteyn, A. & Laserson, U. MHCflurry 2.0: improved pan-allele prediction of MHC class I-presented peptides by incorporating antigen processing. Cell Syst. 11 , 418–419 (2020).

Sarkizova, S. et al. A large peptidome dataset improves HLA class I epitope prediction across most of the human population. Nat. Biotechnol. 38 , 199–209 (2020).

Chen, B. et al. Predicting HLA class II antigen presentation through integrated deep learning. Nat. Biotechnol. 37 , 1332–1343 (2019).

Erhard, F., Dolken, L., Schilling, B. & Schlosser, A. Identification of the cryptic HLA-I immunopeptidome. Cancer Immunol. Res. 8 , 1018–1026 (2020).

Ouspenskaia, T. et al. Unannotated proteins expand the MHC-I-restricted immunopeptidome in cancer. Nat. Biotechnol. 40 , 209–217 (2022).

Blass, E. & Ott, P. A. Advances in the development of personalized neoantigen-based therapeutic cancer vaccines. Nat. Rev. Clin. Oncol. 18 , 215–229 (2021).

Sahin, U. et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 547 , 222–226 (2017).

Hilf, N. et al. Actively personalized vaccination trial for newly diagnosed glioblastoma. Nature 565 , 240–245 (2019).

Finn, O. J. A Believer’s overview of cancer immunosurveillance and immunotherapy. J. Immunol. 200 , 385–391 (2018).

Marty, R. et al. MHC-I genotype restricts the oncogenic mutational landscape. Cell 171 , 1272–1283.e15 (2017).

Marty Pyke, R. et al. Evolutionary pressure against MHC class II binding cancer mutations. Cell 175 , 1991 (2018).

Dersh, D., Holly, J. & Yewdell, J. W. A few good peptides: MHC class I-based cancer immunosurveillance and immunoevasion. Nat. Rev. Immunol. 21 , 116–128 (2021).

Van den Eynden, J., Jimenez-Sanchez, A., Miller, M. L. & Larsson, E. Lack of detectable neoantigen depletion signals in the untreated cancer genome. Nat. Genet. 51 , 1741–1748 (2019).

Claeys, A., Luijts, T., Marchal, K. & Van den Eynden, J. Low immunogenicity of common cancer hot spot mutations resulting in false immunogenic selection signals. PLoS Genet. 17 , e1009368 (2021).

Hoyos, D. et al. Fundamental immune-oncogenicity trade-offs define driver mutation fitness. Nature 606 , 172–179 (2022).

O’Donnell, J. S., Teng, M. W. L. & Smyth, M. J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 16 , 151–167 (2019).

Balachandran, V. P. et al. Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer. Nature 551 , 512–516 (2017).

Luksza, M. et al. A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy. Nature 551 , 517–520 (2017).

Luksza, M. et al. Neoantigen quality predicts immunoediting in survivors of pancreatic cancer. Nature 606 , 389–395 (2022).

Fluckiger, A. et al. Cross-reactivity between tumor MHC class I-restricted antigens and an enterococcal bacteriophage. Science 369 , 936–942 (2020).

Zitvogel, L. & Kroemer, G. Cross-reactivity between cancer and microbial antigens. Oncoimmunology 10 , 1877416 (2021).

Au, L. et al. Determinants of anti-PD-1 response and resistance in clear cell renal cell carcinoma. Cancer Cell 39 , 1497–1518.e11 (2021).

Yarchoan, M., Hopkins, A. & Jaffee, E. M. Tumor mutational burden and response rate to PD-1 inhibition. N. Engl. J. Med. 377 , 2500–2501 (2017).

Devarakonda, S. et al. Tumor mutation burden as a biomarker in resected non-small-cell lung cancer. J. Clin. Oncol. 36 , 2995–3006 (2018).

Samstein, R. M. et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat. Genet. 51 , 202–206 (2019).

Attermann, A. S., Bjerregaard, A. M., Saini, S. K., Gronbaek, K. & Hadrup, S. R. Human endogenous retroviruses and their implication for immunotherapeutics of cancer. Ann. Oncol. 29 , 2183–2191 (2018).

Faulkner, G. J. et al. The regulated retrotransposon transcriptome of mammalian cells. Nat. Genet. 41 , 563–571 (2009).

Rycaj, K. et al. Cytotoxicity of human endogenous retrovirus K-specific T cells toward autologous ovarian cancer cells. Clin. Cancer Res. 21 , 471–483 (2015).

Jang, H. S. et al. Transposable elements drive widespread expression of oncogenes in human cancers. Nat. Genet. 51 , 611–617 (2019).

Smith, C. C. et al. Endogenous retroviral signatures predict immunotherapy response in clear cell renal cell carcinoma. J. Clin. Invest. 128 , 4804–4820 (2018).

Saini, S. K. et al. Human endogenous retroviruses form a reservoir of T cell targets in hematological cancers. Nat. Commun. 11 , 5660 (2020).

Bullman, S. et al. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science 358 , 1443–1448 (2017).

Nejman, D. et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science 368 , 973–980 (2020).

Jin, C. et al. Commensal microbiota promote lung cancer development via γδ T cells. Cell 176 , 998–1013.e16 (2019).

Riquelme, E. et al. Tumor microbiome diversity and composition influence pancreatic cancer outcomes. Cell 178 , 795–806.e12 (2019).

Fu, A. et al. Tumor-resident intracellular microbiota promotes metastatic colonization in breast cancer. Cell 185 , 1356–1372.e26 (2022).

Matson, V. et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 359 , 104–108 (2018).

Routy, B. et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 359 , 91–97 (2018).

Sepich-Poore, G. D., Carter, H. & Knight, R. Intratumoral bacteria generate a new class of therapeutically relevant tumor antigens in melanoma. Cancer Cell 39 , 601–603 (2021).

Xavier, J. B. et al. The cancer microbiome: distinguishing direct and indirect effects requires a systemic view. Trends Cancer 6 , 192–204 (2020).

Yoo, S. K., Chowell, D., Valero, C., Morris, L. G. T. & Chan, T. A. Outcomes among patients with or without obesity and with cancer following treatment with immune checkpoint blockade. JAMA Netw. Open 5 , e220448 (2022).

Yoo, S. K., Chowell, D., Valero, C., Morris, L. G. T. & Chan, T. A. Pre-treatment serum albumin and mutational burden as biomarkers of response to immune checkpoint blockade. NPJ Precis. Oncol. 6 , 23 (2022).

Krishna, C., Chowell, D., Gonen, M., Elhanati, Y. & Chan, T. A. Genetic and environmental determinants of human TCR repertoire diversity. Immun. Ageing 17 , 26 (2020).

Middha, S. et al. Majority of B2M-mutant and -deficient colorectal carcinomas achieve clinical benefit from immune checkpoint inhibitor therapy and are microsatellite instability-high. JCO Precis. Oncol. 3 , PO.18.00321 (2019).

PubMed   PubMed Central   Google Scholar  

Yeon Yeon, S. et al. Immune checkpoint blockade resistance-related B2M hotspot mutations in microsatellite-unstable colorectal carcinoma. Pathol. Res. Pract. 215 , 209–214 (2019).

Zhang, C. et al. B2M and JAK1/2-mutated MSI-H colorectal carcinomas can benefit from anti-PD-1 therapy. J. Immunother. 45 , 187–193 (2022).

Zhang, H. et al. B2M overexpression correlates with malignancy and immune signatures in human gliomas. Sci. Rep. 11 , 5045 (2021).

Li, D. et al. β2-Microglobulin maintains glioblastoma stem cells and induces M2-like polarization of tumor-associated macrophages. Cancer Res. 82 , 3321–3334 (2022).

Bern, M. D. et al. Inducible down-regulation of MHC class I results in natural killer cell tolerance. J. Exp. Med. 216 , 99–116 (2019).

McGranahan, N. et al. Allele-specific HLA loss and immune escape in lung cancer evolution. Cell 171 , 1259–1271.e11 (2017).

Gong, X. & Karchin, R. Pan-cancer HLA gene-mediated tumor immunogenicity and immune evasion. Mol. Cancer Res. 20 , 1272–1283 (2022).

Thorsson, V. et al. The immune landscape of cancer. Immunity 48 , 812–830.e14 (2018).

Shklovskaya, E. et al. Tumor MHC expression guides first-line immunotherapy selection in melanoma. Cancers 12 , 3374 (2020).

Liu, D. et al. Integrative molecular and clinical modeling of clinical outcomes to PD1 blockade in patients with metastatic melanoma. Nat. Med. 25 , 1916–1927 (2019).

Axelrod, M. L., Cook, R. S., Johnson, D. B. & Balko, J. M. Biological consequences of MHC-II expression by tumor cells in cancer. Clin. Cancer Res. 25 , 2392–2402 (2019).

Neuwelt, A. J. et al. Cancer cell-intrinsic expression of MHC II in lung cancer cell lines is actively restricted by MEK/ERK signaling and epigenetic mechanisms. J. Immunother. Cancer 8 , e000441 (2020).

Johnson, D. B. et al. Melanoma-specific MHC-II expression represents a tumour-autonomous phenotype and predicts response to anti-PD-1/PD-L1 therapy. Nat. Commun. 7 , 10582 (2016).

Roemer, M. G. M. et al. Major histocompatibility complex class II and programmed death ligand 1 expression predict outcome after programmed death 1 blockade in classic Hodgkin lymphoma. J. Clin. Oncol. 36 , 942–950 (2018).

Gonzalez-Ericsson, P. I. et al. Tumor-specific major histocompatibility-II expression predicts benefit to anti-PD-1/L1 therapy in patients with HER2-negative primary breast cancer. Clin. Cancer Res. 27 , 5299–5306 (2021).

Schirrmacher, V., Schild, H. J., Guckel, B. & von Hoegen, P. Tumour-specific CTL response requiring interactions of four different cell types and recognition of MHC class I and class II restricted tumour antigens. Immunol. Cell Biol. 71 , 311–326 (1993).

Tay, R. E., Richardson, E. K. & Toh, H. C. Revisiting the role of CD4 + T cells in cancer immunotherapy-new insights into old paradigms. Cancer Gene Ther. 28 , 5–17 (2021).

Zander, R. et al. CD4 + T cell help is required for the formation of a cytolytic CD8 + T cell subset that protects against chronic infection and cancer. Immunity 51 , 1028–1042.e4 (2019).

Rosenthal, R. et al. Neoantigen-directed immune escape in lung cancer evolution. Nature 567 , 479–485 (2019).

Thol, K., Pawlik, P. & McGranahan, N. Therapy sculpts the complex interplay between cancer and the immune system during tumour evolution. Genome Med. 14 , 137 (2022).

Homma, Y. et al. Changes in the immune cell population and cell proliferation in peripheral blood after gemcitabine-based chemotherapy for pancreatic cancer. Clin. Transl. Oncol. 16 , 330–335 (2014).

Jimenez-Sanchez, A. et al. Unraveling tumor-immune heterogeneity in advanced ovarian cancer uncovers immunogenic effect of chemotherapy. Nat. Genet. 52 , 582–593 (2020).

Szikriszt, B. et al. A comprehensive survey of the mutagenic impact of common cancer cytotoxics. Genome Biol. 17 , 99 (2016).

Wu, T. et al. Quantification of neoantigen-mediated immunoediting in cancer evolution. Cancer Res. 82 , 2226–2238 (2022).

Motzer, R. J. et al. Molecular subsets in renal cancer determine outcome to checkpoint and angiogenesis blockade. Cancer Cell 38 , 803–817.e4 (2020).

Dong, L. Q. et al. Heterogeneous immunogenomic features and distinct escape mechanisms in multifocal hepatocellular carcinoma. J. Hepatol. 72 , 896–908 (2020).

Tawbi, H. A. et al. Relatlimab and nivolumab versus nivolumab in untreated advanced melanoma. N. Engl. J. Med. 386 , 24–34 (2022).

Frampton, A. E. & Sivakumar, S. A new combination immunotherapy in advanced melanoma. N. Engl. J. Med. 386 , 91–92 (2022).

Marin-Acevedo, J. A., Kimbrough, E. O. & Lou, Y. Next generation of immune checkpoint inhibitors and beyond. J. Hematol. Oncol. 14 , 45 (2021).

Guo, E. et al. WEE1 inhibition induces anti-tumor immunity by activating ERV and the dsRNA pathway. J. Exp. Med. 219 , e20210789 (2022).

Andre, P. et al. Anti-NKG2A mAb is a checkpoint inhibitor that promotes anti-tumor immunity by unleashing both T and NK cells. Cell 175 , 1731–1743.e13 (2018).

Roschewski, M., Longo, D. L. & Wilson, W. H. CAR T-cell therapy for large B-cell lymphoma — who, when, and how? N. Engl. J. Med. 386 , 692–696 (2022).

Yarmarkovich, M. et al. Cross-HLA targeting of intracellular oncoproteins with peptide-centric CARs. Nature 599 , 477–484 (2021).

Bonaventura, P. et al. Identification of shared tumor epitopes from endogenous retroviruses inducing high-avidity cytotoxic T cells for cancer immunotherapy. Sci. Adv. 8 , eabj3671 (2022).

Rapoport, A. P. et al. NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat. Med. 21 , 914–921 (2015).

Cameron, B. J. et al. Identification of a Titin-derived HLA-A1-presented peptide as a cross-reactive target for engineered MAGE A3-directed T cells. Sci. Transl. Med. 5 , 197ra103 (2013).

Zhao, X. et al. Tuning T cell receptor sensitivity through catch bond engineering. Science 376 , eabl5282 (2022).

Bekes, M., Langley, D. R. & Crews, C. M. PROTAC targeted protein degraders: the past is prologue. Nat. Rev. Drug Discov. 21 , 181–200 (2022).

Massafra, V. et al. Proteolysis-targeting chimeras enhance T cell bispecific antibody-driven T cell activation and effector function through increased MHC class I antigen presentation in cancer cells. J. Immunol. 207 , 493–504 (2021).

Duan, Z. & Ho, M. T-cell receptor mimic antibodies for cancer immunotherapy. Mol. Cancer Ther. 20 , 1533–1541 (2021).

Hansen, T. H., Connolly, J. M., Gould, K. G. & Fremont, D. H. Basic and translational applications of engineered MHC class I proteins. Trends Immunol. 31 , 363–369 (2010).

Chang, A. Y. et al. A therapeutic T cell receptor mimic antibody targets tumor-associated PRAME peptide/HLA-I antigens. J. Clin. Invest. 127 , 3557 (2017).

Dao, T. et al. Targeting the intracellular WT1 oncogene product with a therapeutic human antibody. Sci. Transl. Med. 5 , 176ra133 (2013).

Article   Google Scholar  

Carvajal, R. D. et al. Advances in the clinical management of uveal melanoma. Nat. Rev. Clin. Oncol. 20 , 99–115 (2023).

Nathan, P. et al. Overall survival benefit with tebentafusp in metastatic uveal melanoma. N. Engl. J. Med. 385 , 1196–1206 (2021).

Carvajal, R. D. et al. Phase I study of safety, tolerability, and efficacy of tebentafusp using a step-up dosing regimen and expansion in patients with metastatic uveal melanoma. J. Clin. Oncol. 40 , 1939–1948 (2022).

Liu, X. et al. Development of a TCR-like antibody and chimeric antigen receptor against NY-ESO-1/HLA-A2 for cancer immunotherapy. J. Immunother. Cancer 10 , e004035 (2022).

Hsiue, E. H. et al. Targeting a neoantigen derived from a common TP53 mutation. Science 371 , eabc8697 (2021).

Douglass, J. et al. Bispecific antibodies targeting mutant RAS neoantigens. Sci. Immunol. 6 , eabd5515 (2021).

Ma, J. et al. Bispecific antibodies: from research to clinical application. Front. Immunol. 12 , 626616 (2021).

Hegde, P. S. & Chen, D. S. Top 10 challenges in cancer immunotherapy. Immunity 52 , 17–35 (2020).

FDA. FDA Approves Pembrolizumab for Adults and children with TMB-H Solid Tumors https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-pembrolizumab-adults-and-children-tmb-h-solid-tumors (2020).

FDA. FDA Grants Accelerated Approval to Dostarlimab-gxly for dMMR Advanced Solid Tumors https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-dostarlimab-gxly-dmmr-advanced-solid-tumors (2021).

Valero, C. et al. The association between tumor mutational burden and prognosis is dependent on treatment context. Nat. Genet. 53 , 11–15 (2021).

Valero, C. et al. Response rates to anti-PD-1 immunotherapy in microsatellite-stable solid tumors with 10 or more mutations per megabase. JAMA Oncol. 7 , 739–743 (2021).

Hellmann, M. D. et al. Tumor mutational burden and efficacy of Nivolumab monotherapy and in combination with Ipilimumab in small-cell lung cancer. Cancer Cell 33 , 853–861.e4 (2018).

Wang, F. et al. Evaluation of POLE and POLD1 mutations as biomarkers for immunotherapy outcomes across multiple cancer types. JAMA Oncol. 5 , 1504–1506 (2019).

Andre, T. et al. Pembrolizumab in microsatellite-instability-high advanced colorectal cancer. N. Engl. J. Med. 383 , 2207–2218 (2020).

Le, D. T. et al. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 372 , 2509–2520 (2015).

Ma, X. et al. Functional landscapes of POLE and POLD1 mutations in checkpoint blockade-dependent antitumor immunity. Nat. Genet. 54 , 996–1012 (2022).

Bauer, J. et al. The oncogenic fusion protein DNAJB1-PRKACA can be specifically targeted by peptide-based immunotherapy in fibrolamellar hepatocellular carcinoma. Nat. Commun. 13 , 6401 (2022).

Weber, D. et al. Accurate detection of tumor-specific gene fusions reveals strongly immunogenic personal neo-antigens. Nat. Biotechnol. 40 , 1276–1284 (2022).

Yoshihama, S. et al. NLRC5/CITA expression correlates with efficient response to checkpoint blockade immunotherapy. Sci. Rep. 11 , 3258 (2021).

Takahashi, A. et al. Tyrosine kinase inhibitors stimulate HLA class I expression by augmenting the IFNγ/STAT1 signaling in hepatocellular carcinoma cells. Front. Oncol. 11 , 707473 (2021).

Chang, C. H., Hammer, J., Loh, J. E., Fodor, W. L. & Flavell, R. A. The activation of major histocompatibility complex class I genes by interferon regulatory factor-1 (IRF-1). Immunogenetics 35 , 378–384 (1992).

Kriegsman, B. A. et al. Frequent loss of IRF2 in cancers leads to immune evasion through decreased MHC class I antigen presentation and increased PD-L1 expression. J. Immunol. 203 , 1999–2010 (2019).

Schaafsma, E., Fugle, C. M., Wang, X. & Cheng, C. Pan-cancer association of HLA gene expression with cancer prognosis and immunotherapy efficacy. Br. J. Cancer 125 , 422–432 (2021).

Jung, H. et al. DNA methylation loss promotes immune evasion of tumours with high mutation and copy number load. Nat. Commun. 10 , 4278 (2019).

Zhou, L., Mudianto, T., Ma, X., Riley, R. & Uppaluri, R. Targeting EZH2 enhances antigen presentation, antitumor immunity, and circumvents anti-PD-1 resistance in head and neck cancer. Clin. Cancer Res. 26 , 290–300 (2020).

Gambacorta, V. et al. Integrated multiomic profiling identifies the epigenetic regulator PRC2 as a therapeutic target to counteract leukemia immune escape and relapse. Cancer Discov. 12 , 1449–1461 (2022).

Zheng, J. et al. miR-148a-3p silences the CANX/MHC-I pathway and impairs CD8 + T cell-mediated immune attack in colorectal cancer. FASEB J. 35 , e21776 (2021).

Song, D. et al. Insights into the role of ERp57 in cancer. J. Cancer 12 , 2456–2464 (2021).

Nielsen, M. et al. Coexisting alterations of MHC class I antigen presentation and IFNγ signaling mediate acquired resistance of melanoma to post-PD-1 immunotherapy. Cancer Immunol. Res. 10 , 1254–1262 (2022).

Cromme, F. V. et al. Loss of transporter protein, encoded by the TAP-1 gene, is highly correlated with loss of HLA expression in cervical carcinomas. J. Exp. Med. 179 , 335–340 (1994).

Seliger, B. et al. Reduced membrane major histocompatibility complex class I density and stability in a subset of human renal cell carcinomas with low TAP and LMP expression. Clin. Cancer Res. 2 , 1427–1433 (1996).

CAS   PubMed   Google Scholar  

Kaklamanis, L. et al. Loss of major histocompatibility complex-encoded transporter associated with antigen presentation (TAP) in colorectal cancer. Am. J. Pathol. 145 , 505–509 (1994).

CAS   PubMed   PubMed Central   Google Scholar  

Lopez de Castro, J. A. How ERAP1 and ERAP2 shape the peptidomes of disease-associated MHC-I proteins. Front. Immunol. 9 , 2463 (2018).

Download references

Acknowledgements

The authors are grateful for the support from NIH grants R35CA232097 (T.A.C.), R01CA205426 (T.A.C.), U54CA274513 (T.A.C.) and T32CA094186 (K.Y.), a Young Investigator Award from ASCO Conquer Cancer Foundation (K.Y.), a RSNA Research Resident Grant (K.Y.), and a Cleveland Clinic VeloSano Impact Award (K.Y.).

Author information

Authors and affiliations.

Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA

Kailin Yang, Ahmed Halima & Timothy A. Chan

Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA

Timothy A. Chan

National Center for Regenerative Medicine, Cleveland, OH, USA

Case Comprehensive Cancer Center, Cleveland, OH, USA

You can also search for this author in PubMed   Google Scholar

Contributions

All authors made a substantial contribution to all aspects of the preparation of this manuscript.

Corresponding author

Correspondence to Timothy A. Chan .

Ethics declarations

Competing interests.

T.A.C. is a co-founder of and holds equity in Gritstone Oncology; holds equity in An2H; acknowledges grant funding from An2H, AstraZeneca, Bristol Myers Squibb, Eisai, Illumina and Pfizer; has served as an advisor for An2H, AstraZeneca, Bristol Myers Squibb, Eisai, Illumina and MedImmune; and holds ownership of intellectual property on using tumour mutational burden to predict immunotherapy response, which has been licensed to PGDx. K.Y. and A.H. declare no competing interests.

Peer review

Peer review information.

Nature Reviews Clinical Oncology thanks J. Balko and the other, anonymous, peer reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Cite this article.

Yang, K., Halima, A. & Chan, T.A. Antigen presentation in cancer — mechanisms and clinical implications for immunotherapy. Nat Rev Clin Oncol 20 , 604–623 (2023). https://doi.org/10.1038/s41571-023-00789-4

Download citation

Accepted : 25 May 2023

Published : 16 June 2023

Issue Date : September 2023

DOI : https://doi.org/10.1038/s41571-023-00789-4

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

explain the process of antigen presentation

U.S. flag

An official website of the United States government

The .gov means it's official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you're on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings
  • Browse Titles

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Janeway CA Jr, Travers P, Walport M, et al. Immunobiology: The Immune System in Health and Disease. 5th edition. New York: Garland Science; 2001.

By agreement with the publisher, this book is accessible by the search feature, but cannot be browsed.

Cover of Immunobiology

Immunobiology: The Immune System in Health and Disease. 5th edition.

Chapter 5 antigen presentation to t lymphocytes.

In an adaptive immune response , antigen is recognized by two distinct sets of highly variable receptor molecules—the immunoglobulins that serve as antigen receptors on B cells and the antigen-specific receptors of T cells . As we saw in Chapter 3, T cells recognize only antigens that are displayed on cell surfaces. These antigens may derive from pathogens that replicate within cells, such as viruses or intracellular bacteria , or from pathogens or their products that cells internalize by endocytosis from the extracellular fluid. T cells can detect the presence of intracellular pathogens because infected cells display on their surface peptide fragments derived from the pathogens' proteins. These foreign peptides are delivered to the cell surface by specialized host-cell glycoproteins, the MHC molecules , which are also described in Chapter 3. The MHC glycoproteins are encoded in a large cluster of genes that were first identified by their potent effects on the immune response to transplanted tissues. For that reason, the gene complex was termed the major histocompatibility complex (MHC) . We now know that within this region of the genome, in addition to those genes encoding the MHC molecules themselves, are many genes whose products are involved in the production of the MHC:peptide complexes.

We will begin by discussing the mechanisms of antigen processing and presentation, whereby protein antigens are degraded into peptides inside cells and the peptides are then carried to the cell surface bound to MHC molecules . We will see that the two different classes of MHC molecule, known as MHC class I and MHC class II, deliver peptides from different cellular compartments to the surface of the infected cell. Peptides from the cytosol are bound to MHC class I molecules and are recognized by CD8 T cells , whereas peptides generated in vesicles are bound to MHC class II molecules and recognized by CD4 T cells . The two functional subsets of T cells are thereby activated to initiate the destruction of pathogens resident in these two different cellular compartments. Some CD4 T cells activate naive B cells that have internalized specific antigen, and thus also stimulate the production of antibodies to extracellular pathogens and their products.

In the second part of this chapter we will see that there are several genes for each class of MHC molecule: that is, the MHC is polygenic . Each of these genes has many variants: that is, the MHC is also highly polymorphic. Indeed, the most remarkable feature of the MHC class I and II genes is their genetic variability . MHC polymorphism has a profound effect on antigen recognition by T cells , and the combination of polygeny and polymorphism greatly extends the range of peptides that can be presented to T cells by each individual and each population at risk from an infectious pathogen.

  • The generation of T-cell receptor ligands
  • The major histocompatibility complex and its functions
  • Summary to Chapter 5
  • General references
  • Section references
  • Cite this Page Janeway CA Jr, Travers P, Walport M, et al. Immunobiology: The Immune System in Health and Disease. 5th edition. New York: Garland Science; 2001. Chapter 5, Antigen Presentation to T Lymphocytes.
  • Disable Glossary Links

Related Items in Bookshelf

  • All Textbooks

Recent Activity

  • Antigen Presentation to T Lymphocytes - Immunobiology Antigen Presentation to T Lymphocytes - Immunobiology

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

Connect with NLM

National Library of Medicine 8600 Rockville Pike Bethesda, MD 20894

Web Policies FOIA HHS Vulnerability Disclosure

Help Accessibility Careers

statistics

IMAGES

  1. Antigen Processing and Presentation

    explain the process of antigen presentation

  2. PPT

    explain the process of antigen presentation

  3. Mechanism of antigen presentation upon infection/active vaccination

    explain the process of antigen presentation

  4. | Pathways for antigen presentation. (1) Direct-presentation: Antigen

    explain the process of antigen presentation

  5. Antigen Processing and Presentation

    explain the process of antigen presentation

  6. MHC & Antigen Presentation

    explain the process of antigen presentation

VIDEO

  1. Antigens : Structure and properties

  2. 3.Antigen presentation and MHC restriction

  3. Endogenous Antigen Processing and Presentation-MHC-1

  4. Antigen processing & presentation lecture 7

  5. Lec 8: Antigen processing and presentation

  6. ANTIGEN PROCESSING AND PRESENTATIONS IN HINDI #exogenousantigenpathway @ZOOLOGYNOTES #zoology #vbu

COMMENTS

  1. Antigen Processing and Presentation

    Antigen Processing and Presentation. In order to be capable of engaging the key elements of adaptive immunity (specificity, memory, diversity, self/nonself discrimination), antigens have to be processed and presented to immune cells. Antigen presentation is mediated by MHC class I molecules, and the class II molecules found on the surface of ...

  2. Antigen presentation

    Antigen presentation is a vital immune process that is essential for T cell immune response triggering. Because T cells recognize only fragmented antigens displayed on cell surfaces, antigen processing must occur before the antigen fragment can be recognized by a T-cell receptor. Specifically, the fragment, bound to the major histocompatibility ...

  3. Antigen Processing and Presentation

    This is carried out by Antigen-presenting cells (APCs), the most important of which are dendritic cells, B cells, and macrophages. APCs can digest proteins they encounter and display peptide fragments from them on their surfaces for other immune cells to recognise. This process of antigen presentation allows T cells to "see" what proteins ...

  4. A guide to antigen processing and presentation

    Abstract. Antigen processing and presentation are the cornerstones of adaptive immunity. B cells cannot generate high-affinity antibodies without T cell help. CD4 + T cells, which provide such ...

  5. 20.3E: Antigen-Presenting Cells

    Antigen presentation is a process in the body's immune system by which macrophages, dendritic cells and other cell types capture antigens, then present them to naive T-cells. The basis of adaptive immunity lies in the capacity of immune cells to distinguish between the body's own cells and infectious pathogens. The host's cells express ...

  6. 15.4M: Antigen Presentation

    The process: B cells engulf antigen by receptor-mediated endocytosis; The B cell receptors for antigen (BCRs) are antibodies anchored in the plasma membrane. The affinity of these for an epitope on an antigen may be so high that the B cell can bind and internalize the antigen when it is present in body fluids in concentrations thousands of ...

  7. Antigen-Presenting Cells

    Dendritic cells are immune cells that process antigen material; they are present in the skin (Langerhans cells) and the lining of the nose, lungs, stomach, and intestines. Sometimes a dendritic cell presents on the surface of other cells to induce an immune response, thus functioning as an antigen-presenting cell. Macrophages also function as APCs.

  8. Antigen Presentation

    Antigen Presentation and Major Histocompatibility Complex. Pavel P. Nesmiyanov, in Encyclopedia of Infection and Immunity, 2022 Abstract. Antigen presentation is a process that allows T cells to recognize antigenic epitopes displayed on the surface of an antigen-presenting cell. Antigen presentation involves a sophisticated process of epitope preparation, i.e., processing which involves ...

  9. PDF A guide to antigen processing and presentation

    Step 6: display of MHC molecules at the cell surface. Most of the steps of antigen processing and presentation out-lined so far are constitutive in cells that express MHC. molecules. This has ...

  10. Antigen Presentation

    Antigen Processing and Presentation. In Primer to the Immune Response (Second Edition), 2014. A Overview of Antigen Processing and Presentation. Antigen processing is the complex process by which antigens are produced from macromolecules. Most often, antigen processing refers to the generation of antigenic peptides from proteins.

  11. Antigen processing

    Antigen processing, or the cytosolic pathway, is an immunological process that prepares antigens for presentation to special cells of the immune system called T lymphocytes.It is considered to be a stage of antigen presentation pathways. This process involves two distinct pathways for processing of antigens from an organism's own (self) proteins or intracellular pathogens (e.g. viruses), or ...

  12. PDF PowerPoint Presentation

    PowerPoint Presentation. OBJECTIVES. • Identify and explain the locations and functions of MHC molecules. • Identify and explain antigen processing and presentation. • Identify and explain antigen recognition and T-cell selection. • Identify the characteristics of T-dependent and T-independent antigens (including super antigens).

  13. Antigen-presenting cell

    Antigen presentation stimulates immature T cells to become either mature "cytotoxic" CD8+ cells or mature "helper" CD4+ cells. An antigen-presenting cell (APC) or accessory cell is a cell that displays an antigen bound by major histocompatibility complex (MHC) proteins on its surface; this process is known as antigen presentation. T cells may recognize these complexes using their T cell ...

  14. 12.3B: Antigen-Presenting Cells (APCs)

    Antigen-presenting cells (APCs) include dendritic cells, macrophages, and B-lymphocytes. APCs express both MHC-I and MHC-II molecules and serve two major functions during adaptive immunity: they capture and process antigens for presentation to T-lymphocytes, and they produce signals required for the proliferation and differentiation of lymphocytes.

  15. Antigen processing and presentation: Cytosolic and Endocytic pathway

    Antigen processing and Antigen presentation. Antigen processing is a metabolic process that digests the proteins into peptides which can be displayed on the cell membrane together with a class-I or class-II MHC molecules and recognized by T-cells. Antigen presentation is the process by which certain cell in the body especially antigen ...

  16. Role of the antigen presentation process in the immunization mechanism

    The immune system recognizes the exogenous antigen, initiates the inflammatory response and the subsequent steps leading to the production of specific antibodies by the B cells. 2 In human cells, the antigen presentation process is performed by the MHC I and II, and this mechanism is essential for the cell‐mediated immunity. 3 The MHC I is a ...

  17. Biology, Evolution, and History of Antigen Processing and Presentation

    The three of us were fortunate to have lived through this era as young immunologists, awestruck by each new paper that modified our view of antigen presentation. In this special issue of Immunogenetics, we take you from Jan Klein's 1986 history chapter entitled "The Story" to the modern-day perspectives of antigen processing and presentation.

  18. The Role of Antigen Processing and Presentation in Cancer and the

    The processing and presentation pathway is a multi-step process in the context of the normal turnover of cellular proteins and often starts in the cytoplasm. ... Antigen presentation efficiency is also diminished in human tumors characterized by large chromosomal instability and structural alterations. ... This may also explain why MHCI ...

  19. Antigen Presentation

    Antigen presentation can occur in the lymphoid organs (a process known as 'priming') or in the peripheral tissues following pathogen infection. T cells primed by antigen presentation will become activated, expand in number, and migrate to the infected site where they ensure the elimination of the invading microorganism.

  20. Pathways of Antigen Processing

    Figure 2. Trafficking of antigens for processing and presentation with MHC molecules: basic pathways and exceptions to the "rules". Cytosolic proteins are processed primarily by the action of the proteasome. The short peptides are then transported into the ER by TAP for subsequent assembly with MHC-I molecules.

  21. Antigen presentation in cancer

    This latter process generates an antigen-specific signal that is elicited through the binding of the TCR to the antigen-MHC I complex — commonly referred to as signal 1 in the immunology ...

  22. MHC Class I, Class II, Antigen Processing, And Presentation

    Antigen Processing and Presentation. The recognition of protein antigens by T-lymphocytes required that the antigens be processed by Antigen-presenting Cells, then displayed within the cleft of the MHC molecules on the membrane of the cell. This involves the degradation of the protein antigens into peptides, a process known as antigen processing.

  23. Antigen Presentation to T Lymphocytes

    Chapter 5. Antigen Presentation to T Lymphocytes. In an adaptive immune response, antigen is recognized by two distinct sets of highly variable receptor molecules—the immunoglobulins that serve as antigen receptors on B cells and the antigen-specific receptors of T cells. As we saw in Chapter 3, T cells recognize only antigens that are ...