Rheumatoid Arthritis Flashcards

1
Q

What is arthritis, and who is more likely to get it?

A

Arthritis is inflammation of the joints and surrounding tissues, affecting bones, muscles, and joints. It’s an umbrella term for over 100 different conditions. It’s more common in the elderly, women, and individuals with increased weight.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Describe the primary manifestation of rheumatoid arthritis (RA).

A

The primary manifestation of RA is synovitis, which involves inflammation of the synovial lining of joints, leading to erosion of bone, cartilage, and periarticular structures.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What are the articular clinical features of RA?

A

Include morning stiffness of ≥30 minutes, pain, swelling, fatigue, weakness, and characteristic joint deformities like boutonniere, swan neck, Z thumb, MCP subluxation, and ulnar deviation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What are the extra-articular clinical features of RA?

A

Include nodules (subcutaneous, pulmonary, cardiac), pulmonary complications (interstitial lung disease, pleuritis), ocular manifestations (scleritis, episcleritis), vasculitis, neurological issues, cardiovascular complications, cutaneous manifestations, hematologic abnormalities, malignancies, and renal complications.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

How is pyoderma gangrenosum present in patients with RA?

A

Pyoderma gangrenosum in RA starts as a small pimple, red bump, or blood blister, which rapidly enlarges into a painful ulcer with purple or blue edges. It commonly occurs on the legs and heals after treatment with medications like methotrexate and high-dose oral prednisolone.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What are the differences in joint involvement between rheumatoid arthritis (RA) and osteoarthritis (OA) according to Goldman’s Cecil Medicine?

A

RA typically shows symmetric joint involvement affecting the wrist, MCP, and PIP joints, with soft tissue swelling. OA, on the other hand, tends to exhibit asymmetric joint involvement affecting the DIP joints and the base of the thumb, with no soft tissue swelling.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are some of the extra-articular manifestations of rheumatoid arthritis (RA)?

A

Extra-articular manifestations of RA include nodules (subcutaneous, pulmonary, cardiac), pulmonary complications (interstitial lung disease, pleuritis), ocular manifestations (scleritis, episcleritis), vasculitis, neurological issues, cardiovascular complications, cutaneous manifestations, hematologic abnormalities, malignancies, and renal complications.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

How does RA differ from osteoarthritis (OA) in terms of joint involvement

A

RA typically involves symmetric joint inflammation, affecting the wrist, metacarpophalangeal (MCP), and proximal interphalangeal (PIP) joints with soft tissue swelling. In contrast, OA tends to involve asymmetric joint inflammation, affecting distal interphalangeal (DIP) joints and the base of the thumb, with marginal osteophytes and joint space narrowing.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Explain the prevalence and burden of arthritis in Australia.

A

In Australia, around 3.7 million people have arthritis, with prevalence increasing with age and being higher in females, individuals with increased waist circumference, and First Nations populations. Arthritis accounts for 13% of the total burden of disease, with significant healthcare expenditure and socioeconomic costs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What are some of the radiographic features of rheumatoid arthritis (RA) in the wrist?

A

Typical radiographic features of RA in the wrist include osseous erosion at sites like the triquetrum, pisiform, scaphoid, distal radius, ulnar styloid process, and radiocarpal joint. Diffuse cartilage loss and erosion are also observed.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Describe the clinical presentation and symptoms of atlantoaxial (AA) subluxation in RA.

A

AA subluxation in RA can lead to symptoms such as pain radiating up to the occiput, slowly progressive spastic quadriparesis, transient medullary dysfunction (altered conscious state, syncope, loss of sphincter control, dysphagia, vertigo), and neurological deficits.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Describe synovium

A

A thin membrane lining the joint capsule in diarthrodial joints, extending from skeletal tissue and covering the joint cavity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Describe cell types and their normal function

A
  • Cell Types: It consists of synoviocytes, mainly fibroblast-like synoviocytes (FLS) and macrophages.
  • Normal Function: Synoviocytes facilitate joint movement, with FLS producing lubricants like hyaluronan and collagen, while macrophages clean debris and maintain immune balance.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Describe the changes that happen to healthy synovial tissue in RA.

A
  • In RA, synovial tissue undergoes proliferation, infiltration by inflammatory cells (neutrophils, lymphocytes), and neovascularization.
  • Proliferation: Synovial lining layer thickens, forming pannus.
  • Infiltration: Inflammatory cells invade, leading to inflammation and joint damage.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Describe the cell types present and their functions within the inflamed synovium in RA.

A
  • Cell Types: In RA synovium, various cells are present, including activated fibroblast-like synoviocytes (FLS), inflammatory macrophages, T cells, and B cells.
  • Functions: FLS produce pro-inflammatory cytokines and enzymes causing cartilage destruction, while macrophages clean debris and initiate immune responses.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Explain what TNF is what it does in RA

A

A cytokine produced by various cells in RA synovium, promoting inflammation, leukocyte accumulation, and bone resorption.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Explain IL-1 and what is does in RA.

A

Pro-inflammatory cytokine inducing chemokine production, metalloproteinase release, and osteoclast differentiation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Explain what IL-6 is what it does in RA

A

Stimulates acute phase response, immunoglobulin production, and promotes TH17 cell differentiation, contributing to inflammation and bone resorption.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Explain how JAK-STAT signalling contributes to synovial inflammation.

A
  • JAK-STAT Signalling: Janus Kinase (JAK) and Signal Transducer and Activator of Transcription (STAT) pathways mediate inflammatory responses in RA.
  • Contribution: Activated by cytokines like IL-6, JAK-STAT signalling enhances cytokine expression, promotes T cell survival, and induces antibody production, exacerbating synovial inflammation and joint damage.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Explain why animal models are used for pre-clinical studies of RA pathogenesis and treatment.

A
  • Purpose: Animal models, like rodent CIA and hTNF.Tg mice, mimic RA pathogenesis, aiding in understanding disease mechanisms and testing potential therapies.
  • Advantages: They allow controlled studies, testing various interventions, and evaluating treatment responses before human trials.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What are two pre-clinical models of RA

A
  1. Collagen-Induced Arthritis (CIA)
  2. hTNF.Tg mice
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Describe CIA and how it has helped understand RA and its limitations

A
  • Utility: Mimics synovial inflammation, cartilage, and bone erosion seen in RA.
  • Limitations: Dependent on specific mouse strains and collagen antibodies, not perfectly representing human RA.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Describe hTNF.Tg and how it has helped understand RA and its limitations

A
  • Utility: Offers chronic, TNF-driven arthritis model, reflecting RA’s chronic nature and responsiveness to TNF inhibition.
  • Limitations: Limited to TNF-driven mechanisms, not reflecting the multifactorial nature of human RA.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Describe how TNF was identified and developed as a therapeutic target for RA treatment

A
  • Identification: TNF, discovered as a factor killing fibrosarcoma cells, was later found elevated in RA synovium, driving inflammation and joint damage.
  • Development: Monoclonal antibodies like Infliximab and Adalimumab were developed, targeting TNF and reducing synovial inflammation and bone erosion in RA patients.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Describe the anatomical and molecular structure of bone

A

Bone comprises trabecular (spongy) and cortical structures. Trabecular bone is found at the ends of long bones and in the vertebrae, while cortical bone forms the outer shell of most bones. Bone contains bone marrow cells, adipocytes, blood vessels, and extracellular components such as osteoid (organic matrix) and mineral salts (hydroxyapatite).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

Describe the function of bone

A

Bones provide mechanical support for movement and protection for internal organs. They also serve as a metabolic store for minerals like calcium and phosphate, regulate hematopoiesis, and produce hormones and cytokines.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Explain the process of bone remodelling

A

Bone remodeling involves the coordinated action of osteoclasts, which resorb bone, and osteoblasts, which form bone. It is initiated asynchronously throughout the skeleton and is essential for skeletal adaptation and renewal.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Explain how bone remodelling contributes to bone homeostasis

A

Remodeling removes regions of micro-damage, responds to calcium homeostatic needs by releasing calcium into the bloodstream, and adapts to changes in mechanical needs by redistributing bone. It helps maintain bone density and strength.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Describe osteoclasts and how they resorb bone

A

Osteoclasts are multinucleated cells derived from hematopoietic precursors. They attach to bone surfaces and resorb bone mineral and collagenous matrix. Osteoclasts create an acidified microenvironment that aids in bone resorption and possess a characteristic “ruffled border” to increase surface area for ion exchange.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

Describe osteoblasts and how they form bone

A

Osteoblasts are specialized stromal cells responsible for bone formation. They produce type I collagen (osteoid) and non-collagenous proteins, which form the organic matrix of bone. Osteoblasts mineralize this matrix, leading to the formation of new bone.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

Describe osteocytes and their role in the skeleton

A

Osteocytes are terminally differentiated osteoblasts embedded within the bone matrix. They act as mechanosensors, detecting changes in mechanical load, and regulating osteoblast and osteoclast activity accordingly. Osteocytes play a crucial role in bone remodeling and adaptation to mechanical stress.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

Explain the RANKL / OPG system in osteoclast differentiation

A

RANKL (Receptor Activator of Nuclear Factor Kappa-B Ligand) produced by osteoblast-lineage cells stimulates osteoclast formation, while OPG (Osteoprotegerin) inhibits RANKL’s action by acting as a decoy receptor. This system regulates osteoclast differentiation and activity.

33
Q

Explain how the RANKL / OPG system in osteoclast differentiation is regulated in RA.

A

In rheumatoid arthritis (RA), there is an overproduction of RANKL, leading to excessive osteoclast activity and bone resorption. Cytokines such as TNF, IL-1, and IL-6 promote osteoclast differentiation and bone destruction indirectly by inducing RANKL expression in various cell types within the inflamed joint.

34
Q

Define focal bone loss in RA and explain how osteoclasts contribute.

A

Focal bone loss in RA refers to bone erosion within the arthritic joint, particularly at the junction of cartilage and bone. Osteoclasts contribute to focal bone loss by resorbing cortical bone within the affected joint.

35
Q

Define systemic bone loss in RA and explain how osteoclasts contribute.

A

Systemic bone loss in RA involves thinning of trabecular and cortical bone at sites remote from affected joints, increasing susceptibility to fractures. Osteoclasts contribute to systemic bone loss by releasing cytokines into the bloodstream, leading to increased RANKL production and bone resorption throughout the skeleton.

36
Q

Describe how anti-resorptive therapies protect bone in RA.

A

Anti-resorptive therapies in RA aim to inhibit osteoclast activity and bone resorption. Examples include bisphosphonates, which induce osteoclast apoptosis, and denosumab, a RANKL antibody that blocks osteoclast formation. By reducing bone resorption, these therapies protect against bone damage and loss associated with RA.

37
Q

Describe how the inflamed synovium, via TNF, IL-1 and IL-6, causes bone destruction in RA.

A

In RA, the inflamed synovium produces pro-inflammatory cytokines such as TNF, IL-1, and IL-6. These cytokines promote osteoclast differentiation and bone destruction indirectly by inducing RANKL expression in synovial fibroblasts, T cells, and osteoblast-lineage cells.

38
Q

What are the risk factors associated with rheumatoid arthritis (RA)?

A

Risk factors for RA include genetic predisposition (HLA-DRB1 alleles), epigenetic modifications, hormonal influences (e.g., estrogen), environmental triggers (e.g., smoking, infections), and emerging factors such as microbiome alterations.

39
Q

How does genetic susceptibility contribute to the development of RA?

A

Genetic factors, particularly HLA-DRB1 alleles like *0401 and *0404, play a significant role in RA susceptibility. The presence of the shared epitope (SE) within these alleles increases the risk of developing RA and is associated with more severe disease outcomes.

40
Q

What is the role of citrullination in the pathogenesis of RA?

A

Citrullination, a post-translational modification catalyzed by PADI enzymes, leads to the generation of neoepitopes, triggering autoimmunity in RA. Citrullinated peptides bind more avidly to the shared epitope, contributing to the production of anti-citrullinated protein antibodies (ACPA) and proinflammatory cytokines.

41
Q

How do epigenetic mechanisms contribute to RA?

A

Epigenetic modifications, including histone modifications, DNA methylation, and microRNA expression, can alter gene expression without changing DNA sequences. In RA, these mechanisms affect synovitis, immune cell proliferation, and inflammation, influencing disease development and progression.

42
Q

What hormonal factors are associated with RA, particularly in females?

A

Female preponderance in RA suggests a potential role for hormones like estrogen. Estrogen affects immune function, apoptosis resistance in B cells, and production of inflammatory mediators by synoviocytes and macrophages. Pregnancy is associated with temporary remission, likely due to hormonal changes.

43
Q

How do environmental factors such as smoking contribute to RA risk?

A

Smoking is a potent environmental risk factor for RA, particularly in individuals with genetic susceptibility (e.g., HLA-DRB1 alleles). It increases the risk of developing RA and accelerates disease progression by promoting citrullination and immune dysregulation.

44
Q

What emerging theories propose additional mechanisms underlying RA pathogenesis?

A

Emerging theories suggest the involvement of the microbiome and altered vascular permeability in RA. Dysbiosis in the gut microbiota and altered vascular integrity may contribute to immune dysregulation and joint inflammation in susceptible individuals.

45
Q

How can lifestyle modifications help prevent RA?

A

Lifestyle modifications, including smoking cessation, maintaining a healthy weight, regular physical activity, good dental hygiene, breastfeeding, and dietary changes (e.g., omega-3 fatty acids), may reduce the risk of RA by addressing behavioral risk factors and promoting overall health.

46
Q

What autoantibodies are used in the diagnosis of RA, and how do they contribute to disease classification and prognosis

A

Rheumatoid Factor (RF) and Anti-Citrullinated Protein Antibodies (ACPA) are key autoantibodies used in RA diagnosis. Their presence helps classify RA and predict disease progression, severity, and extra-articular manifestations, providing valuable prognostic information.

47
Q

How is disease activity assessed in RA patients, and what are the components of disease activity measurement?

A

Disease activity in RA is assessed through joint counts (tender/swollen), global assessment, pain scores, morning stiffness duration, laboratory tests (ESR, CRP), disability assessment, fatigue evaluation, and radiological imaging to monitor erosive changes. These components help guide treatment decisions and evaluate therapeutic response.

48
Q

Risk factors for rheumatoid arthritis (RA): Genetic

A

Genetic predisposition, particularly HLA-DRB1 alleles like *0401 and *0404, contributes significantly to RA susceptibility. The presence of the shared epitope within these alleles increases the risk of developing RA and is associated with more severe disease outcomes.

49
Q

Risk factors for rheumatoid arthritis (RA): Epigenetic

A

Epigenetic modifications, including histone modifications, DNA methylation, and microRNA expression, can alter gene expression without changing DNA sequences. These mechanisms affect synovitis, immune cell proliferation, and inflammation, influencing RA development and progression.

50
Q

Risk factors for rheumatoid arthritis (RA): Environmental

A

Environmental factors such as smoking, infections, and exposure to pollutants play a significant role in RA development. Smoking, in particular, is a potent risk factor that accelerates disease progression by promoting citrullination and immune dysregulation.

51
Q

Risk factors for rheumatoid arthritis (RA): Hormonal

A

Female preponderance in RA suggests a potential role for hormones like estrogen. Estrogen affects immune function, apoptosis resistance in B cells, and production of inflammatory mediators by synoviocytes and macrophages. Pregnancy is associated with temporary remission, likely due to hormonal changes.

52
Q

Risk factors for rheumatoid arthritis (RA): Emerging factors

A

Emerging theories propose additional mechanisms underlying RA pathogenesis, including dysbiosis in the gut microbiota and altered vascular integrity. These factors may contribute to immune dysregulation and joint inflammation in susceptible individuals.

53
Q

Risk factors for rheumatoid arthritis (RA): Preventing RA

A

Lifestyle modifications, including smoking cessation, maintaining a healthy weight, regular physical activity, good dental hygiene, breastfeeding, and dietary changes (e.g., omega-3 fatty acids), may reduce the risk of RA by addressing behavioral risk factors and promoting overall health.

54
Q

Diagnosis: Role of autoantibodies

A

Autoantibodies, particularly Rheumatoid Factor (RF) and Anti-Citrullinated Protein Antibodies (ACPA), play a crucial role in the diagnosis of RA. Their presence helps classify RA and predict disease progression, severity, and extra-articular manifestations, providing valuable prognostic information.

55
Q

Diagnosis: Classification criteria

A

The 2010 ACR/EULAR classification criteria for RA help identify patients who should be tested for autoantibodies and diagnosed with RA. These criteria include the presence of definite clinical synovitis in one or more joints and synovitis not better explained by another disease. Sensitivity and specificity of these criteria are essential for accurate diagnosis and treatment initiation.

56
Q

What are the principles of rheumatoid arthritis (RA) treatment regarding the timing of intervention?

A

RA treatment should begin early, within the first three months of symptom onset, during the therapeutic “window of opportunity.”

57
Q

Describe the therapeutic approach recommended for RA management.

A

The therapeutic approach for RA emphasizes early treatment initiation, aiming for remission or low disease activity through a treat-to-target strategy, and employing intensive therapeutic regimens.

58
Q

What is the significance of the “window of opportunity” in RA treatment?

A

The “window of opportunity” refers to the early phase of RA when intervention can significantly impact disease progression, reduce chronicity, and minimize joint damage, leading to improved long-term outcomes.

59
Q

What medications are used in RA treatment, and what are the categories of disease-modifying anti-rheumatic drugs (DMARDs)?

A

RA treatment includes analgesics, non-steroidal anti-inflammatory drugs (NSAIDs), glucocorticoids, conventional synthetic DMARDs (csDMARDs), biological DMARDs (bDMARDs), and targeted synthetic DMARDs (tsDMARDs). DMARDs are categorized into csDMARDs and bDMARDs.

60
Q

How are conventional synthetic DMARDs (csDMARDs) typically initiated in RA management?

A

After an RA diagnosis, csDMARDs, such as methotrexate, are usually started early, either as monotherapy or in combination therapy, with methotrexate being the first-line choice for the majority of patients.

61
Q

What evidence supports the use of intensive therapeutic regimens in RA management?

A

Studies like TICORA (tight intensive control of RA) and triple therapy trials provide evidence for the effectiveness of intensive therapeutic regimens, emphasizing the importance of tight disease control and combination therapy.

62
Q

How are biological DMARDs (bDMARDs) typically used in RA treatment, and what are the available classes?

A

bDMARDs are reserved for RA patients who do not respond adequately to csDMARDs. There are five classes of bDMARDs: TNF-α inhibitors, IL-1 antagonists, IL-6 receptor antagonists, CTLA4 ligands, and B-cell depleting agents.

63
Q

Explain the eligibility criteria for receiving bDMARDs under the Pharmaceutical Benefits Scheme (PBS) in Australia.

A

To be eligible for bDMARDs under the PBS, patients must have failed intensive csDMARD therapy, meet specific disease activity criteria (ESR, CRP, and active joint count), and fulfill other clinical requirements.

63
Q

What is the role of the PBS in providing access to medications for RA patients in Australia?

A

The PBS subsidizes the cost of medications for various medical conditions, including RA, ensuring affordable access to necessary treatments for patients.

64
Q

Summarise the key principles of RA treatment and the significance of early intervention.

A

RA treatment involves early initiation of therapy within the “window of opportunity,” employing a treat-to-target approach with intensive regimens. Medications include csDMARDs and bDMARDs, with PBS criteria guiding access to subsidized treatments in Australia.

65
Q

What are the principles of RA treatment?

A

The principles include treating early and aiming for a target of low disease activity or remission.

66
Q

Define the “window of opportunity” in RA treatment.

A

The “window of opportunity” refers to the first three months after symptom onset, during which early intervention may significantly reduce disease progression and the burden of RA.

67
Q

What is the main goal of Phase I clinical trials?

A

Phase I trials aim to evaluate the safety of a new intervention, typically involving a small group of people to assess pharmacokinetics, pharmacodynamics, and toxicity.

68
Q

What is the primary purpose of Phase III clinical trials?

A

Phase III trials compare the new intervention to the current standard of treatment in large groups of patients to confirm efficacy and safety.

69
Q

List the classes of biological DMARDs (bDMARDs) for RA treatment.

A
  1. Tumor necrosis factor alpha (TNF-α) inhibitors
  2. Interleukin one (IL-1) antagonists
  3. Interleukin six (IL-6) receptor antagonists
  4. Cytotoxic T-lymphocyte antigen 4 (CTLA4) ligand (co-stimulation modifier)
  5. B-cell depleting agents (anti-CD20)
70
Q

What are the eligibility criteria for bDMARDs under the Pharmaceutical Benefits Scheme (PBS) in Australia?

A

Patients must have failed six months of intensive DMARD therapy, with specific clinical criteria such as active joint counts and elevated inflammatory markers.

71
Q

Describe the mechanism of action of Methotrexate (MTX) in RA treatment.

A

Methotrexate is the first-line csDMARD choice for RA and is believed to exert its effects through mechanisms that are not fully understood. It is typically used as an anchor drug to which other medications may be added.

72
Q

What are the common adverse effects associated with TNF inhibitors in RA treatment?

A

Common adverse effects include injection site reactions, infusion reactions, cytopenias (neutropenia), and increased susceptibility to infections.

73
Q

What is the American College of Rheumatology 20% improvement criteria (ACR20)?

A

ACR20 is used to evaluate treatment response in clinical trials and requires at least a 20% improvement in swollen and tender joint counts, along with improvements in additional disease-related variables.

74
Q

Name three emerging therapies for RA treatment.

A

Mavrilimumab (GM-CSF inhibition), Rheumavax (autologous dendritic cell therapy), and cholinergic neurostimulation are emerging therapies being explored for RA treatment.

75
Q

Human Clinical Trials of TNFi:

A

Human clinical trials of TNFi (Tumor Necrosis Factor inhibitors) have demonstrated their efficacy in the treatment of various autoimmune diseases, including rheumatoid arthritis (RA). These trials typically involve different phases, starting from Phase I trials to evaluate safety and escalating to Phase III trials to assess efficacy in larger patient populations.

76
Q

Principles of Clinical Trial Design:

A
  1. Ethical considerations: Trials must adhere to ethical guidelines to ensure the safety and well-being of participants.
  2. Randomization: Random assignment of participants to treatment groups helps minimize bias and ensures comparability between groups.
  3. Placebo/control: Utilization of placebos or active controls allows researchers to assess the true effect of the intervention being studied.
  4. Blinding (masking): Blinding helps prevent bias by ensuring that participants and/or researchers are unaware of which treatment is being administered.
  5. Adequate power (sample size): Trials need to enroll a sufficient number of participants to detect clinically meaningful differences between treatment groups.
77
Q

TNF-Inhibitors (TNFi):

A

TNFi comprises different classes of biologic agents that target tumor necrosis factor-alpha (TNF-α), a pro-inflammatory cytokine implicated in various autoimmune diseases. These classes include:

  • Monoclonal anti-TNF antibodies: Examples include Infliximab, Adalimumab, Certolizumab pegol, and Golimumab.
  • TNF receptor fusion proteins: Etanercept is an example of this class.
  • Efficacy: TNFi have demonstrated efficacy in reducing disease activity, improving symptoms, and inhibiting joint damage in RA and other autoimmune conditions.
  • Failure: Despite their efficacy, some patients may experience primary or secondary failure with TNFi therapy. Primary failure occurs when patients do not respond to TNFi due to factors such as different inflammatory pathways driving the disease. Secondary failure may occur over time due to the development of neutralizing antibodies or other mechanisms leading to loss of efficacy.
78
Q

Future of RA Treatment:

A

The future of RA treatment is evolving with the development of new therapeutic agents and treatment paradigms. Some aspects of the future of RA treatment include:

  1. Evolving treatment paradigms: There is a shift towards “head-to-head” trials comparing different biologic DMARDs (bDMARDs) and biosimilars to assess their relative efficacy and safety.
  2. Introduction of biosimilars: Biosimilars, which are lower-cost alternatives to biologic drugs, are becoming increasingly available, potentially reducing the economic burden of RA treatment.
  3. New therapeutic agents: Small molecule inhibitors targeting Janus Kinase (JAK) pathways, such as tofacitinib, baricitinib, and upadacitinib, offer alternative treatment options for RA patients.
  4. Emerging therapies: Novel agents targeting pathways involved in RA pathogenesis, such as GM-CSF inhibition and cholinergic neurostimulation, are being investigated for their potential in RA treatment.