25 ธันวาคม 2553

ยาที่ใช้ในการรักษา SLE : Tacrolimus

Tacrolimus

ที่มา  : ปริ่มเฉนียน มุ่งการดี.ตำรายาใหม่ในประเทศไทยเล่ม 9 : คลังข้อมูลยา มหาวิทยาลัยมหิดล.


FIGURE detail in the Sites of action of available immunosuppressants that inhibit the three-signal model of T-cell activation and proliferation.  From the following article:Individuality: the barrier to optimal immunosuppression Barry D.    KahanNature Reviews Immunology 3, 831-838 (October 2003)




 




22 ธันวาคม 2553

Guidance for Industry Systemic Lupus Erythematosus — Developing Medical Products for Treatment.

 สำหรับบทความนี้จะ เป็นภาษาอังกฤษทั้งหมดเลยนะคะ อาจจะเข้าใจยากหน่อย แต่อย่างไรก็ตาม เจ้าของบล็อคไม่กล้าแปลเนื่องจากกลัวความผิดพลาดในการแปลเกิดขึ้น 


เป็นการกำหนดข้อกำหนดต่างๆ เกี่ยวกับโรงงานที่ต้องการผลิต ยา อุปกรณ์ใดๆ เกี่ยวกับการใช้ในผู้ป่วยที่มีโรค SLE ว่าจะต้องเป็นไปตามมาตรฐานนี้ ของ FDA หรือองค์การอนามัยโลก ตามประกาศนี้ เท่านั้น ยังไงลองอ่านดูได้นะคะ ว่ามีการประกาศข้อกำหนดอะไรไว้บ้าง

From : U.S. Department of Health and Human Services
Food and Drug Administration
Center for Drug Evaluation and Research (CDER)
Center for Biologics Evaluation and Research (CBER)
Center for Devices and Radiological Health (CDRH)
June 2010
Clinical/Medical

This guidance represents the Food and Drug Administration’s (FDA’s) current thinking on this topic.
It  does not create or confer any rights for or on any person and does not operate to bind FDA or the public.
You can use an alternative approach if the approach satisfies the requirements of the applicable statutes  and regulations.  If you want to discuss an alternative approach, contact the FDA staff responsible for implementing this guidance.
If you cannot identify the appropriate FDA staff, call the appropriate number listed on the title page of this guidance.

I. INTRODUCTION
The purpose of this guidance is to assist sponsors in the clinical development of medical
products (i.e., human drugs, therapeutic biological products, and medical devices) for the
treatment of systemic lupus erythematosus (SLE).  Specifically, this guidance addresses the Food
and Drug Administration’s (FDA’s) current thinking regarding the overall development program
and clinical trial designs, and provides specific information on trial design, trial duration,
efficacy endpoints, and response criteria.  This guidance is intended to serve as a focus for
continued discussions among the FDA, medical industry, sponsors, academic community, and
the public.
2
  As the science of this indication evolves, this guidance may be revised.  
This guidance applies to general information regarding medical product development for the
treatment of SLE.  Organ-specific forms of disease will be addressed in separate guidances.
This guidance does not contain discussion of the general issues of clinical trial design or
statistical analysis.  Those topics are addressed in the ICH guidances for industry E9 Statistical
Principles for Clinical Trials and E10 Choice of Control Group and Related Issues in Clinical
                                               
1
 This guidance has been prepared by the Systemic Lupus Erythematosus Working Group, which includes
representatives from the Center for Drug Evaluation and Research (CDER), the Center for Biologics Evaluation and
Research (CBER), the Center for Devices and Radiological Health (CDRH), and the Office of Critical Path
Programs (OCPP) in the Office of the Commissioner (OC) at the Food and Drug Administration. 
2
 In addition to consulting guidances, sponsors are encouraged to contact the relevant division to discuss specific
issues that arise during the development of SLE medical products. 
1Contains Nonbinding Recommendations
Trials.
3
  This guidance focuses on specific medical product development and trial design issues
that are unique to the study of SLE.
FDA’s guidance documents, including this guidance, do not establish legally enforceable
responsibilities.  Instead, guidances describe the Agency’s current thinking on a topic and should
be viewed only as recommendations, unless specific regulatory or statutory requirements are
cited.  The use of the word should in Agency guidances means that something is suggested or
recommended, but not required.
II. BACKGROUND
SLE is a chronic disease characterized by protean manifestations, often with a waxing and
waning course.  In the past, a diagnosis of SLE often implied a decreased life span caused by
internal organ system involvement or the toxic effects of therapy, but recent improvements in
care have dramatically enhanced the survival of SLE patients.  Nonetheless, increased mortality
remains a major concern and current treatments for SLE remain inadequate.
4
  Many patients
have incompletely controlled disease, progression to end-stage organ involvement continues, and
the therapies carry risks of debilitating side effects.  Therefore, it is important to facilitate the
development of medical products that have the potential to be more effective and/or less toxic.  
III. DEVELOPMENT PROGRAM
A. General Considerations
1. Early Phase Clinical Development Considerations
Studies to identify an appropriate (safe and effective) dose are an important component of phase
2 development for human drugs and therapeutic biological products used to treat SLE.  For
additional information on the FDA’s current thinking regarding exposure response or dose
response, see the ICH guidance for industry E4 Dose-Response Information to Support Drug
Registration and the guidances for industry Providing Clinical Evidence of Effectiveness for
Human Drug and Biological Products and Exposure-Response Relationships — Study Design,
Data Analysis, and Regulatory Applications.  
We recommend that early studies evaluate concurrent use of a new medical product with
commonly used standard therapies to obtain preliminary safety information on potential
interactions with medical products used in standard-of-care regimens, although at this stage
studies will not be powered to fully assess safety endpoints.  As discussed in section III.B.8.,
Primary Efficacy Endpoints, early exploratory clinical studies can be used to gain experience
                                               
3
 We update guidances periodically.  To make sure you have the most recent version of a guidance, check the FDA
Guidances Web page at http://www.fda.gov/RegulatoryInformation/Guidances/default.htm.
4
 Ippolito, A and M Petri, 2008, An Update on Mortality in Systemic Lupus Erythematosus, Clin Exp Rheumatol,
26(5 Suppl 51):S72-9.
2Contains Nonbinding Recommendations
with a variety of standard clinical outcome measures, which may aid sponsors in determining
which endpoints to pursue further in phase 3 trials. 
Biomarker assays thought to reflect disease activity also can be helpful in identifying medical
products likely to show a clinical benefit and in choosing doses and regimens.  See section
III.B.10., Other Endpoints, for additional information on the use of biomarkers in SLE clinical
studies.
2. Efficacy Considerations
The evidence of effectiveness needed to support approval of medical products for SLE is similar
to that for medical products for other indications.  For human drugs and therapeutic biological
products, at least two adequate and well-controlled trials generally are needed for approval. 
However, a single study can suffice under some circumstances(see the guidance for industry
Providing Clinical Evidence of Effectiveness for Human Drugs and Biological Products).  A
single study can be sufficient, for example, if the medical product is being developed for the
treatment of serious acute manifestations and the study shows:  (1) robust evidence of efficacy
with resolution of the serious acute manifestations; or (2) a decrease in mortality.  For medical
devices, one confirmatory clinical trial generally is sufficient.  
B. Specific Efficacy Trial Considerations
1. Indication
The general indication of treatment of systemic lupus erythematosus will be granted for medical
products if supported by sufficient evidence of effectiveness.  In general, specific efficacy claims
(e.g., reduction in disease activity), as discussed in section III.B.8., Primary Efficacy Endpoints,
will not be included in the INDICATIONS AND USAGE section of labeling, but can be
discussed in the CLINICAL STUDIES section if well-supported.  If the medical product is
studied only in a subset of the general SLE population, then the restricted population in which
the medical product was studied would be reflected in labeling.  For medical products that
demonstrate a reduction in mortality in adequate and well-controlled trials, appropriate additional
labeling reflecting that outcome would be included in the INDICATIONS AND USAGE section.
2. Study Design
a. Superiority trials
The preferred design for efficacy trials is a parallel, randomized, controlled superiority trial using
placebo or active control.  The placebo-controlled trial can compare the test medical product
with no treatment, but more commonly adds the test medical product or placebo to standard
therapy (add-on trial).  
No patient enrolling in an SLE clinical trial should be denied standard therapy if doing so would
lead to irreversible harm.  To avoid denying patients standard of care, superiority trials of new
therapies can use an add-on design, if the medical product is intended as adjunctive treatment, or
3Contains Nonbinding Recommendations
head-to-head comparisons with an alternative standard of care, if the medical product is intended
for primary treatment.  In a head-to-head comparison, it may be appropriate to include early
escape provisions to an alternative standard-of-care regimen for patients who worsen during the
study to ensure that no patient is denied potentially effective therapy.
One of the advantages to an add-on trial of this type is that it enables the evaluation of drug
effects in the context of commonly used medical products in SLE.  An example of an add-on
design would be a trial of corticosteroids plus placebo compared to corticosteroids plus the new
medical product.  The protocol should specify the dose of corticosteroids patients will receive,
taking into account the type and severity of the clinical manifestation.  The protocol also should
include provisions for tapering of corticosteroids during the trial if the manifestations improve.
b. Noninferiority trials
If superiority to a comparator is unlikely (e.g., because the new medical product is
pharmacologically similar to a standard-of-care medical product) and an add-on study would be
unlikely to succeed (again because the new and standard-of-care medical products are
pharmacologically similar), sponsors might want to consider a noninferiority design to evaluate
efficacy.  However, this design would be difficult to support in this case (see ICH E10).  To use a
noninferiority design, the effect size of the comparator that will be present in the new study must
be identified to define the noninferiority margin.
5
  Currently, there are no known medical
products with an effect size adequately characterized to design an adequate noninferiority trial
for a new medical product in any SLE setting.  A particular problem would also be the inherent
variability in outcome and response in different populations.  Sponsors considering a
noninferiority design should discuss the design with the appropriate review division before trial
initiation.
c. Extension trials
If prior evidence suggests clinical activity of the medical product and an acceptable safety
profile, sponsors are encouraged to offer patients enrollment into a long-term extension trial to
characterize long-term safety.  Long-term controlled trial data are preferred over open-label
extension safety data because of the difficulty in interpreting adverse event rates in the absence
of a concurrent control.  Demonstration of long-term benefit would be a critical determination in
some settings (e.g., bone marrow transplant).
d. Alternative trial designs
Alternative trial designs, all of which should be designed as superiority trials, include
randomized withdrawal, dose-response, and replacement trials.  In a replacement trial, patients
on a stable standard-of-care regimen should be randomized in a blinded manner to continue that
regimen or switch to study treatment.  A successful trial would demonstrate better outcomes in
the group switched to study treatment.  Sponsors should discuss these alternative designs with
the review division before initiating these studies.
                                               
5
 See 21 CFR 314.126.
4Contains Nonbinding Recommendations
3. Study Duration
Clinical trials should be of sufficient length to assess the durability of therapy benefits, taking
into account the chronic nature of SLE and its waxing and waning course.  In general, a trial
evaluating the following endpoints should be at least 1 year in duration:  reduction in disease
activity, complete clinical response or remission, reduction in flare/increase in time to flare, and
maintenance of response.  The duration should be based on the onset of action of the medical
product and incorporate maintenance therapy for a total of at least 1 year to assess for durability
of response as well as safety, depending upon the risks of the medical product.   
If the investigational medical product is intended for short-term use, such as induction of
response, the total duration of follow-up should still be 1 year, but the investigational medical
product does not need to be continued beyond the initial treatment period.  In this case, patients
can be switched to another maintenance therapy for the remainder of the follow-up period.
Studies investigating treatment of serious acute manifestations of SLE (e.g., acute confusional
state, acute transverse myelitis, or acute lupus pneumonitis) are considered a special case of
induction therapy.  Such studies can also be of relatively short duration depending on the nature
of the manifestation, the organ system involved, and the expected time for resolution of the
serious acute manifestations under investigation.  As for any other trial of induction therapy, a
subsequent assessment of the durability of response and safety should be based on data of at least
1-year duration.  
4. Study Population
Trials should enroll patients with established SLE, as defined by the American College of
Rheumatology classification criteria.  
The patient population should reflect the patients who would reasonably be considered for the
therapy should it be shown to be effective.  It is important that the studied population be one that
can be generalized to an appropriate population for recommended use, and not made artificially
narrow.  However, if existing data (e.g., from exploratory studies) suggest that only a specific,
limited population can be expected to benefit from the therapy, the inclusion and exclusion
criteria can limit enrollment to that subset of patients with a particular range of disease activity or
with a particular serious acute manifestation of SLE.  However, as discussed in section III.B.1.,
Indication, the medical product, if approved, would be labeled to indicate this restricted
population. 
5. Concomitant Medications
It is important to recognize that changes in concomitant medications, whether steroids,
immunosuppressive agents, or other therapies (e.g., angiotensin converting enzyme inhibitors,
antihypertensive agents, and agents to control diabetes), can influence outcomes and confound
the effect of treatment.  Treating physicians should respond to patient needs appropriately, but an
attempt should be made in the protocol to define the baseline therapy that is acceptable and
5Contains Nonbinding Recommendations
provide guidance on how therapy should be adjusted.  Sponsors should collect complete
information on use of concomitant medications during trials.   
It is important that investigators consider restricting baseline glucocorticoid use (e.g., stable dose
or limit the range of doses) to reduce the variability of dosing that may make interpretation of
results more difficult.  The protocol should specify if glucocorticoid dose changes are allowed
during the trial or if patients should be discontinued if they require an increased glucocorticoid
dosage.
Potential eligible patients should not be deliberately tapered off their concomitant medications to
induce a flare in disease activity for purposes of meeting enrollment criteria in a trial.
We also recommend defining the use of rescue medications and specifying how patients needing
such treatment will be treated and analyzed.  
6. Stratification
If the effects of treatment are expected to differ substantially in patients with severely active
disease as compared to moderately or mildly active disease, it may be desirable to stratify at
randomization.  
The Systemic Lupus Erythematosus International Collaborating Clinics/American College of
Rheumatology (SLICC/ACR) Damage Index was developed, and found to be well-defined and
reliable, for measuring organ damage.  It may be particularly useful as a means of stratifying
patients at trial entry because increased damage has been shown to correlate with a worse
prognostic outcome.
6
If it is expected that particular demographic groups may respond differently to therapy, sponsors
also can consider stratification based on a demographic variable.
7. Pediatric Populations
To help standardize the conduct and reporting of pediatric SLE clinical trials and enhance
identification of new medical products, the Pediatric Rheumatology International Trials
Organization, in collaboration with the Pediatric Rheumatology Collaborative Study Group and
with the support of the European Union and the National Institutes of Health, has developed a
core set of five domains for the evaluation of response to therapy.  These domains include the
following: 
1. A disease activity index (DAI) (e.g., European Consensus Lupus Activity Measure
(ECLAM), Systemic Lupus Erythematosus Disease Activity Index (SLEDAI), Systemic
Lupus Erythematosus Activity Measure (SLAM), British Isles Lupus Assessment Group
(BILAG), or other DAI deemed appropriate for clinical trials) 
                                               
6
 Stoll, T, B Seifert, and DA Isenberg, 1996, SLICC/ACR Damage Index Is Valid, and Renal and Pulmonary Organ
Scores Are Predictors of Severe Outcome in Patients with SLE, Br. J. Rheumatol, 35:248-54.
6Contains Nonbinding Recommendations
2. Renal function (24-hour proteinuria) 
3. Parent’s global 
4. Physician’s global 
5. Health status (Child Health Questionnaire physical summary score) 
Evaluation of response in these five domains can be considered for exploratory use in pediatric
SLE trials.  Future research will help to establish the degree of change in these domains that
represents a clinically important benefit to establish efficacy in clinical trials.  Sponsors should
discuss the design of pediatric SLE trials with the review division before beginning such trials.
8. Primary Efficacy Endpoints
Sponsors should consider designing clinical trials for medical products to address one or more of
the following primary endpoints, as discussed in detail below. 
a. Reduction in disease activity 
The primary endpoint for a trial evaluating reduction in signs and symptoms of SLE disease
activity can be determined using a DAI that has documented evidence of validity, reliability, and
ability to detect change in the targeted clinical trial setting.  Disease activity scores allow
inclusion of patients whose disease affects different organ systems by providing an overall
severity score.  
Disease activity should be measured at the beginning and end of the trial as well as over the
course of the trial.  To meet the primary endpoint of the trial, the change in DAI between the
outset and the end of the trial should show a statistically significant difference between the
treatment groups.  It is also important to determine that an improvement in DAI score is not
accompanied by a worsening of other disease manifestations.  (See also section III.B.14., RiskBenefit Considerations.)
Several indices exist that mirror the assessment of experienced clinicians and are sensitive to
changes in disease activity.  The BILAG is the preferred index to study reduction in disease
activity in clinical trials.  The BILAG scores patients based on the need for therapy; therefore,
the clinical interpretation of a change in score is apparent.  
Other DAIs include the SLEDAI and Safety of Estrogen in Lupus Erythematosus National
Assessment Trial (SELENA)-SLEDAI, the SLAM, and the ECLAM.  Updated versions of the
BILAG, SLAM, and SLEDAI have been released (BILAG2004, SELENA-SLEDAI/SLEDAI
2K, and SLAM-R).  These indices have been shown to be valid in some treatment settings based
on the concordance of scores with expert opinion, acceptable interobserver variability among
trained evaluators, correlation between individual patient scores on different indices, and
correlation between increases in scores and clinical decisions to increase therapy.  They also
have been shown in cohort studies to be sensitive to changes in disease activity, and can be used
7Contains Nonbinding Recommendations
in clinical trials if the instrument measurement properties are adequate for the specific clinical
trial setting.
7
  
It is important to ensure that the selected DAIs accurately assess disease activity over time. 
Some DAIs allot points for a new disease manifestation and no points for a stable manifestation. 
Thus, a disease manifestation that is present at screening and that is stable during the trial can
contribute points to the baseline score but no points to subsequent scores leading to an artifactual
reduction in the overall disease activity score.  The DAIs should also address disease
manifestations not caused by SLE and how they will be scored (e.g., hematuria and/or pyuria
caused by a urinary tract infection versus lupus nephritis).
If using the BILAG in a 1-year SLE trial, sponsors should conduct an assessment of disease
activity at both 6 months and 12 months, as well as at other time points (e.g., monthly) to assess
the time course of response.  The timing of the primary efficacy analysis, at either 6 or 12
months, depends upon the time it takes for the new medical product to achieve optimal activity. 
If 12 months is chosen as the primary endpoint, BILAG should show a statistically significant
improvement at 12 months that has been sustained at a minimum for 2 months.  Alternatively, if
the primary endpoint is set at 6 months, clinical benefit should be assessed at 12 months as a
secondary endpoint.  
In patients with active disease at baseline (defined as one or more BILAG A or two or more
BILAG B scores), the primary efficacy analysis using a clinically meaningful benefit can be
based on the outcome of major clinical response (MCR) or partial clinical response (PCR),
showing a greater frequency in drug-treated patients than in control-treated patients.  
An example of the definition of MCR or PCR is presented here.  In the example of PCR, flare is
defined as the presence of one or more new BILAG A scores or two or more new BILAG B
scores.  BILAG C scores do not affect the definition of flare, since, by definition, they are not
judged to be serious enough to require treatment.
An adjudication committee should be employed to determine which patients meet the predefined
outcome.  The following factors can be used to define MCR and PCR:
Major Clinical Response
A patient with BILAG C scores or better at 6 months with no new BILAG A or
BILAG B scores AND maintenance of response with no new BILAG A or B
scores between 6 and 12 months.
Partial Clinical Response
A patient with BILAG C scores or better at 6 months with no new BILAG A or
BILAG B scores and maintenance of response without a flare for 4 months. 
                                               
7
 Strand, V, D Gladman, D Isenberg, M Petri, J Smolen, and P Tugwell, 1999, Outcome Measures to Be Used in
Clinical Trials in Systemic Lupus Erythematosus, J Rheumatol, 26(2):490-7.
8Contains Nonbinding Recommendations
OR
A patient with a maximum of 1 BILAG B score or better at 6 months AND
maintenance of response without a flare out to 1 year.
OR
A patient with very high disease activity (as defined below) who achieves a
maximum of 2 BILAG B scores at 6 months AND maintenance of this response
without developing a flare out to 1 year.  Very high disease activity is defined as
the presence of one of the following conditions: 
• ≥ 2 BILAG A scores (regardless of the number of BILAG B scores)
OR 
• 1 BILAG A score and ≥ 2 BILAG B scores 
OR
• ≥ 4 BILAG B scores (with no BILAG A scores)
A trial of a new medical product’s ability to induce response in patients with active disease can
also be conducted using a DAI, such as BILAG.  In this case, the primary endpoint would be an
increase in the proportion of patients with a category C score or better at the end of induction
(e.g., 3 or 6 months).  Response should be confirmed by repeat measurement at least 1 month
later.  It is also important that a new medical product not only demonstrate early activity, but also
not worsen long-term outcome.  Therefore, the maintenance of response also should be assessed
as a secondary endpoint at 1 year.
Some treatments may target a biologic mechanism that leads to only certain disease
manifestations, or to only disease manifestations related to a single organ system.  In these
situations, it would usually be preferable to use an organ-specific measure of disease activity as
the primary endpoint as opposed to an overall disease activity measure.  
The interpretation of a clinical trial using the organ-specific approach can be problematic,
however, if improvement in the organ system selected is counterbalanced by worsening
manifestations of disease occurring in other organ systems.  In addition, results from organspecific trials may be confounded if changes in treatment regimens are made, such as an increase
in immunosuppressive agents (see section III.B.5., Concomitant Medications).  Therefore, organspecific trials should also assess overall disease activity as a secondary endpoint, because the
safety information should be taken into consideration in determining the overall risk-benefit
assessment of the medical product. 
b. Complete clinical response or remission
The primary endpoint for a trial evaluating complete clinical response or remission is defined by
the complete absence of disease activity, using a DAI (as described above).  The term response is
used if the patients continue to receive SLE-directed therapies, whereas remission is used if
patients do not continue to receive ongoing therapy for SLE.  
9Contains Nonbinding Recommendations
The evaluation of efficacy should be based on the proportion of patients who achieve a BILAG
level D score, or zero if using the SLEDAI, in all organ systems for at least 6 consecutive
months.   
c. Reduction in flare/increase in time to flare
The primary endpoint for a trial evaluating flares can be a reduction in flares or an increase in the
time to flare for the new medical product compared to the control group.  If time to flare is
evaluated as the primary endpoint, the trial should be at least 1 year in duration to evaluate
whether the flares are suppressed or only delayed in occurrence.  A critical secondary endpoint
should be comparison of flare rates or proportion of patients flare-free at an appropriate time
point.  
A trial assessing flares should randomize patients with quiescent disease (e.g., BILAG C score or
better in all organ systems) and assess flares in the group receiving the new medical product
compared to the control group.  
The definition of flare should be specified in the protocol and should reflect an episode of
increased disease activity that correlates with the need for an increase in or change in treatment
on clinical grounds.  All possible flares should be adjudicated by a data monitoring board that is
blinded to treatment.  
An index used to measure flare should measure disease activity over a month’s period, rather
than at fixed time points, in order not to miss any intercurrent flares and to allow full
characterization of activity of the medical product over the course of the trial.  Acceptable flare
indices for clinical trials include the BILAG and SELENA-SLEDAI flare indexes.  For example,
the BILAG identifies flares as A for severe flare and B for mild to moderate flare.  A worsening
from an E, D, or C to two or more B scores or one or more A score in any body or organ system
during the 1-year trial period can be used to define the occurrence of flare.  Time to flare should
be the number of days since randomization to occurrence of flare based on BILAG A or B
scores.
A trial evaluating maintenance of response can also be considered for a new medical product
once active disease (i.e., flares) is in remission (defined as BILAG C or better).  Such trials can
be a continuation of an induction trial of the new medical product.  Patients with active disease
who achieve quiescence following induction with a new medical product can be further
randomized to switch to placebo or continue the new medical product for the duration of the
trial.  Alternatively, the induction regimen can consist of a standard-of-care regimen whereby
patients are randomized to continue standard of care or are switched to the new medical product
for maintenance.  The primary endpoint for a trial evaluating maintenance of response can be
met by demonstrating an increase (compared to standard of care) in the proportion of patients
maintaining a BILAG C score or better at 1 year.  If the endpoint is assessed at 6 months, then
clinical benefit should also be assessed at 1 year as a secondary endpoint.  
10Contains Nonbinding Recommendations
d. Reduction in concomitant steroids
Reducing corticosteroid use is an important goal in treatment of patients with SLE if it occurs in
the context of a treatment that effectively controls disease activity.  Therefore, for a medical
product to be labeled as reducing corticosteroid usage, it should also demonstrate another clinical
benefit, such as reduction in disease activity as the primary endpoint.  
In an add-on trial to test the steroid-sparing potential of a new medical product, patients should
be enrolled during a flare and randomized to the addition of the new medical product or placebo
to induction doses of corticosteroids.  In both study arms, when patients achieve quiescent
disease, the corticosteroid dose should be tapered to a maintenance dose that is not usually
associated with major toxicities while still maintaining quiescence.  The induction steroid dosage
and duration of induction therapy and taper schedule should be based on the severity of disease
activity in the dominant organ system involved.
8
The evaluation of efficacy should be based on the proportion of patients in treatment and control
groups that achieve a reduction in steroid dose to less than or equal to 10 mg per day of
prednisone or equivalent, with quiescent disease and no flares (see definition above) for at least 3
consecutive months during a 1-year clinical trial.  For a result to be clinically meaningful, the
patient population should be on moderate to high doses of steroids at baseline.  Trials should also
assess the occurrence of clinically significant steroid toxicities. 
e. Treatment of serious acute manifestations
Treatment of serious acute manifestations of SLE can be considered a special case of induction
therapy for treatment of SLE emergencies (e.g., acute confusional state, acute transverse
myelitis, or acute lupus pneumonitis).  The primary endpoints for a trial evaluating treatment of
serious acute manifestations should reflect the proportion of patients with improvement after
administration of a new medical product or placebo.  The improvement should be measured as a
lower score in the organ system score on a DAI of the involved organ(s), such that there is no
longer a threat to that organ.  
As stated in section III.B.3., Study Duration, studies investigating treatment of serious acute
manifestations of SLE are considered a special case of induction therapy.  Therefore, therapy
with the investigational medical product can be of relatively short duration depending on the
nature of the manifestation.  It is understood that in many cases maintenance therapy will involve
a different regimen than the study drug used for induction.  Assessment of the durability of
response and safety should be obtained after the patient is switched to maintenance therapy for a
total of at least 1-year duration.  
Trials investigating treatments for serious acute manifestations of SLE can include the following
secondary endpoints:  time to resolution of the acute manifestation, mortality, need for retreatment, use of corticosteroids, and overall disease activity as measured by a DAI, such as the
SLEDAI or BILAG. 
                                               
8
 Ad Hoc Working Group on Steroid-Sparing Criteria in Lupus, 2004, Criteria for Steroid-Sparing Ability of
Interventions in Systemic Lupus Erythematosus:  Report of a Consensus Meeting, Arthritis & Rheum, 50:3427.
11Contains Nonbinding Recommendations
Study designs investigating therapies for serious acute manifestations of SLE should be
discussed with the review division before beginning trials.  
9. Patient-Reported Outcomes
We recognize that improvements in clinical outcome measures (e.g., lab tests, clinical
evaluation) in patients with SLE may not always translate to improvements in how patients feel
or function.  Therefore, we encourage the use of patient-reported outcome (PRO) instruments to
measure all relevant and important SLE symptoms and patient-perceived abilities to function and
perform daily activities.  PRO instrument development should be based upon qualitative research
conducted in the target patient population to ensure the content validity of the measure.
Most experts agree that fatigue is an important symptom in SLE.  However, experts and patients
define fatigue differently.  Measurement of fatigue in SLE should include the following:  (1) a
clear definition of fatigue as it relates to patients with SLE; (2) a clear conceptual framework
describing fatigue in SLE including physical and mental components, as appropriate; and (3)
methods for measuring fatigue symptoms and effect in the presence of comorbid factors (e.g.,
depression and medication effects).  We have not identified an existing PRO instrument optimal
for measurement of fatigue symptom complex in patients with SLE to support labeling claims. 
Therefore, an exploratory endpoint measure consisting of the use of an existing fatigue measure
as well as an open-ended item that asks patients to identify their symptoms could be useful in the
development of future instruments for measuring fatigue in SLE.  
PRO instruments should be used as key secondary endpoints in all SLE trials.  PRO instruments
that are intended as key trial endpoints should be demonstrated to be well-defined and reliable in
the SLE trial population.  We encourage development of new PRO instruments where
appropriate.  Additional information on how the FDA reviews PRO instruments used to support
medical product labeling can be found in the guidance for industry Patient-Reported Outcome
Measures:  Use in Medical Product Development to Support Labeling Claims. 
10. Other Endpoints
Endpoints other than those discussed above for consideration in particular SLE trials are
discussed here.
a. Damage
An assessment of damage caused by manifestations of SLE disease should be considered for
inclusion in SLE trials of at least 1-year duration.
Use of the SLICC/ACR Damage Index measures irreversible organ system damage caused by
SLE disease that has been present for at least 6 months.  The SLICC/ACR Damage Index
assesses damage that accrues over time in the renal, pulmonary, cardiovascular, and other organ
systems.  It can be used in clinical trials to measure the rate of progression of damage caused by
12Contains Nonbinding Recommendations
the disease, or its treatment, but is not sensitive to change unless the time interval for observation
is at least 1 year in duration.  
An assessment of damage during a trial also can be complicated if a new therapy is associated
with toxicities not measured by the Damage Index (e.g., in organs not associated with SLE
disease).  Therefore, we recommend discussing use of the SLICC/ACR Damage Index or other
instrument to assess damage with the review division before beginning trials.  
b. Biomarkers
A biomarker is a characteristic that is objectively measured and evaluated as an indicator of
normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic
intervention.  In some cases, biomarkers can assist in the development and evaluation of
therapies for SLE by supporting a hypothesized mechanism of action or by suggesting an
appropriate dose or duration of action.  
Surrogate endpoints are a subset of biomarkers that are expected to predict clinical benefit (or
harm or lack of benefit) and are intended to substitute for a clinical endpoint.
9
  Currently, none
of the known biomarkers (e.g., anti-dsDNA levels, complement levels) in SLE has been
validated as a surrogate endpoint, and therefore no biomarker can substitute for a direct
assessment of clinical benefit in clinical trials. 
                                               
In some cases, biomarkers are used to define risk or identify potential responders to a treatment. 
Sponsors should consult the appropriate FDA center to determine whether a biomarker used to
select patients or monitor response in clinical trials can be used in prescribing the medical
product if it is approved (e.g., for selection of patients or for monitoring safety or
effectiveness).
10
11. Study Procedures and Timing of Assessments
In SLE trials using reduction in disease activity as an endpoint, it is important that the protocol
specify procedures to ensure that the scoring of the DAI specifically reflects SLE-related organ
dysfunction.  The interpretation of score changes can be confounded if organ system dysfunction
caused by a disease or condition other than SLE is present or organ dysfunction caused by the
treatment occurs.  Investigators should be appropriately trained to ensure uniform scoring, as
variability can decrease study power.  In some cases, it may be helpful to have an adjudication
committee confirm assessment based on DAIs (e.g., flares or quiescence of disease).
12. Statistical Considerations
The particular statistical analysis used can differ depending on the endpoints and outcomes of
interest.  To assess a reduction in disease activity or flare, induction of response, treatment of
9
 Biomarkers Definitions Working Group, 2001, Biomarkers and Surrogate Endpoints:  Preferred Definitions and
Conceptual Framework, Clin Pharm Therap, 69(3):89-95.
10
 Contact CBER or CDRH’s Office of In Vitro Diagnostic Devices Evaluation and Safety.
13Contains Nonbinding Recommendations
serious acute manifestations, or maintenance of response, the statistical test usually should
evaluate the difference between the treatment and control groups in the proportion of patients
meeting a predefined outcome, although measures of continuous variety also can be useful. 
These outcomes can be summarized as binary or ordinal for the purpose of the primary analysis. 
Although outcomes at the end of the trial are usually the primary focus, outcomes also should be
evaluated at multiple times during the trial.  To assess the time to flare in patients with quiescent
disease, the statistical test usually would evaluate the difference in the time-to-event curves using
an appropriate test.  This analysis also should be supported by an analysis comparing the
proportion of flare-free patients at the end of the trial.  Analysis considerations of primary
endpoints for organ-specific disease should be similar to those for SLE. 
In addition to the primary assessments of disease activity, other aspects of the disease process
may be important in fully elucidating the effect of the treatment on patients.  The overall
probability of a false positive finding for a completely ineffective treatment should be controlled
by prespecifying a single primary analysis or several analyses with appropriate adjustment for
multiplicity.  Secondary analyses also should be adjusted to avoid error and the protocol should
describe the plan for controlling such errors.
We recommend prespecifying in the protocol statistical approaches (e.g., regarding dropouts or
missing data) (see ICH E9).
13. Accelerated Approval Considerations for Human Drugs and Therapeutic
Biological Products (Subpart H and Subpart E)
For serious or life-threatening conditions, a new human drug (21 CFR part 314, subpart H) or
therapeutic biological product (21 CFR part 601, subpart E) can be approved on the basis of
adequate and well-controlled clinical trials that establish that the human drug or therapeutic
biological product has an effect on a “surrogate endpoint that is reasonably likely, based on
epidemiologic, therapeutic, pathophysiologic, or other evidence, to predict clinical benefit” (21
CFR 314.510 and 21 CFR 601.40).  Full approval would be contingent on required
postmarketing clinical trials to verify the clinical benefit.   
No surrogate marker has been reliably shown to predict clinical benefit in patients with SLE and
there has been no Subpart H or Subpart E approval of medical products for SLE.  Sponsors
should be very cautious about selecting a potential surrogate marker intended to support
accelerated approval until there is confidence regarding its predictive value.  
14. Risk-Benefit Considerations
Assessment of risks and benefits involves an appraisal of the effect of the medical product on all
aspects of the disease process, including disease activity, irreversible damage caused by the
disease or its treatment, and health-related quality of life.
11
  The primary efficacy analysis should
show a statistically significant result and the measured clinical effect of the medical product
should be clinically meaningful.  Toxicities related to the pharmacologic effects of the medical
                                               
11
 Strand, V, D Gladman, D Isenberg, et al., 1999, Outcome Measures to Be Used in Clinical Trials in Systemic
Lupus Erythematosus, J Rheumatol, 26:490-7.
14Contains Nonbinding Recommendations
15
                                               
product (e.g., immunosuppression) also should be considered as part of this overall risk-benefit
assessment of the medical product.
It is important that the size of the safety database for human drugs and therapeutic biological
products at approval be consistent with the recommendations made in the ICH guidance for
industry E1A The Extent of Population Exposure to Assess Clinical Safety:  For Drugs Intended
for Long-Term Treatment of Non-Life-Threatening Conditions.  Particular attention should be
paid to the assessment of known toxicities, or to pharmacologic and biological effects that might
be suspected to imply delayed toxicities.  It is important to consider these toxicities in
formulating the clinical development program.  This information may influence the size of the
safety database.  
A smaller safety database may be appropriate to support approval of medical products designed
to treat aspects of SLE that represent orphan indications or for the treatment of serious acute
manifestations, because it may be impossible or impractical to study a large number of patients
with these conditions.
12
  Sponsors may wish to discuss these issues with the appropriate review
division early in the development of a new treatment.  
Finally, if there is concern about rare but serious adverse events (e.g., from the mechanism of
action or experience with similar human drugs and therapeutic biological products), a
postmarketing study or clinical trial may be needed to gather additional safety information. 
12
 For information regarding orphan indications, see the following Web site: 
http://www.fda.gov/ForIndustry/DevelopingProductsforRareDiseasesConditions/HowtoapplyforOrphanProductDesi
gnation/ucm135122.htm

SLE patients with renal damage incur higher health care costs.

SLE patients with renal damage incur higher health care costs


  1. 1Department of Medicine, Division of Clinical Immunology/Allergy, 2Department of Medicine, Division of Clinical Epidemiology, McGill University Health Centre, McGill University, Montréal, Québec, 3Department of Medicine, Division of Rheumatology, University of California, San Francisco, CA, 4Johns Hopkins University School of Medicine, Baltimore, MD, 5Department of Medicine, Division of Rheumatology, and 6Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA, 7Centre for Rheumatology, Department of Medicine, University College London, London, 8Department of Rheumatology, Division of Immunity and Infection, University of Birmingham, Birmingham, UK, 9Department of Medicine, Division of Rheumatology, Centre Hospitalier de L’Université de Montréal, Université de Montréal, 10Department of Epidemiology and Biostatistics, McGill University, Montréal, Québec and 11Global Epidemiology and Outcomes Research, Bristol-Myers Squibb Company, Princeton, NJ, USA.
  1. Correspondence to: A. Clarke, McGill University Health Centre (MUHC), 687 Pine Avenue West, V Building, Montreal, Quebec, Canada H3A 1A1.
  • Received August 28, 2007.
  • Revision received December 17, 2007

จากการศึกษาของคณะที่มีรายชื่อข้างต้น โดยมี วัตถุประสงค์ เพื่อเปรียบเทียบต้นทุนและคุณภาพชีวิต (คุณภาพชีวิต) ในผู้ป่วยโรค SLE ที่มีและไม่มีความเสียหายของไต

วิธีการศึกษาได้ทำโดย ทำการศึกษาในผู้ป่วย เจ็ดร้อยสิบห้าคน โดยทำการสัมภาษณ์ในผู้ป่วยที่มีโรคตั้งแต่ครึ่งปีขึ้นไปจนถึงที่ป่วยมาเป็นระยะเวลา 4 ปี เพื่อสืบค้นหาค่าใช้จ่ายในการใช้การดูแลสุขภาพและการสูญเสียรายได้รวมถึงคุณภาพชีวิตที่เปลีย่นแปลงไป โดยทำเป็นประจำทุกปี


ในส่วนของ ค่าใช้จ่ายตรงและทางอ้อมพบว่า มีค่าใช้จ่ายสะสมเพิ่มขึ้นเป็น (2006 เหรียญแคนาดา) และส่วนของคุณภาพชีวิต (เปลี่ยนเฉลี่ยรายปีใน SF - 36) โดยมาเปรียบเทียบระหว่างผู้ป่วยที่มีและไม่มีความเสียหายของไต [Lupus โดยการวิจัยทำอย่างเป็นระบบมีการทำงานร่วมกันทางคลินิกระหว่างประเทศ/ ACR ความเสียหายดัชนี (SLICC / ACR DI)] ของทั้งสองสภาวะร่วมกัน

ผลการศึกษาสรุปออกมาว่า การศึกษาสำหรับผู้ป่วยที่มีอาการแทรกซ้อนของโรคไต  พบว่า   subscale ของ SLICC / ACR DI = 0 (n = 634), 1(n = 54), 2 (n = 15) และ 3 (n = 12) ค่าเฉลี่ย 4 ปีสะสม ค่าใช้จ่ายโดยตรงต่อผู้ป่วย (95% CI) เท่ากับ 20 $ 337 (18 $ 815, $ 21 858), 27 $ 869 (19 $ 230, $ 36 509), 51 $ 191 (23 $463, $ 78 919) และ 99 $ 544 (57 $ 102, $ 141 987) ตามลำดับ   ในสมการถดถอยที่subscale ไตของ SLICC / ACR DI เป็นตัวบ่งชี้ตัวแปรเดียวโดยเฉลี่ย (95% CI) เพิ่มขึ้นในแต่ละปีที่หน่วยไตเกิดความเสียหาย  โดยเพิ่มขึ้นในอัตราส่วนโดยตรงที่ 24% (15%, 33%) ของค่าใช้จ่ายที่เำพิ่มขึ้น .   สามารถแสดงโดยใช้สมการถดถอยในระดับที่ subscale โดยที่ความเสียหายเป็นตัวแปรที่สามารถบ่งชี้ว่าผู้ป่วยที่มีโรคไตวายเรื้อรังระยะสุดท้าย จะมีค่าใช้จ่ายในการรักษาที่เกิดขึ้นเพิ่มขึ้นไปถึงที่ 103% (65%, 141%) สูงกว่ากว่าของผู้ป่วยที่ไม่มีความเสียหายของไต    แต่ในส่วนของค่าใช้จ่ายทางอ้อมสะสมและการเปลี่ยนแปลงรายปีใน SF - 36คะแนนสรุปได้ว่า ค่าใช้จ่ายทางอ้อมไม่แตกต่างกันระหว่างผู้ป่วยทั้งสองกลุ่ม สรุปผลการวิจัยกล่าวคือ
  ผู้ป่วยโรคลูปัส (SLE) ที่มีความผิดปกติที่ลุกลามไปจนถึงการทำให้เกิดความเสียหายที่ไตสูง จะเกิดค่าใช้จ่ายในการรักษาตัวโดยตรงที่สูงกว่าค่าใช้จ่ายทางอ้อม แต่ในส่วนของคุณภาพชีวิตนั้น พบว่า ทั้งสองกลุ่มไม่มีความแตกต่างอย่างมีนัยสำคัญ ทั้งนี้ หากผู้ป่วยที่มีโรค SLE ที่มีฐานะยากจนอยู่แล้ว อาจมีผลกระทบที่มากกว่านี้ก็ได้ โดยจะเพิม่ขึ้นตามความเสื่อมของไต
ทั้งนี้ เมื่อเวลาผ่านไป ผู้ป่วยก็จะสามารถเปลี่ยนแปลงและยอมรับสภาพและสามารถใช้ชีวิตได้อย่างคนปกติได้ ถึงแม้ว่าอาจจะมีความเสื่อมของไตเกิดขึ้นมากขึ้นเรื่อยๆก็ตาม
ทีี่มา : SLE Treatment updated 2007 โดย :    1. โดย  : A. E. Clarke P. Panopalis3M. Petri 4. S. Manzi  5. D. A. Isenberg6. C. Gordon  7. J.-L. Senécal8. L. Joseph 9. Y. St Pierre2 and 10. T. Li11

 

 

16 ธันวาคม 2553

การแพ้ยา

การแพ้ยา

เมื่อคุณรับประทานยาแล้วเกิดผื่น หรือแน่นหน้าอก แสดงว่าคุณอาจจะมีอาการแพ้ยา แต่การเกิดผลข้างเคียงจากยามิใช่หมายความว่าแพ้ยาเสมอไป อาจจะเกิดจากสาเหตุอื่นก็ได้

และผู้ป่วยก็ยังสามารถรับยานั้นได้ แต่ถ้าหากเกิดจากแพ้ยาผู้ป่วยต้องหลีกเลี่ยงยาที่แพ้โดยเด็ดขาด
การแพ้ยาหมายถึงเกิดปฏิกิริยาภูมิแพ้มักจะเกิดอาการหลังรับประทานยา ทันทีหรือไม่เกิน 2 ชั่วโมงอาการที่สำคัญได้แก่

􀂷 ผื่นคัน
􀂷 คัดจมูก
􀂷 หายใจไม่ออก หายใจเสี่ยงดังหวีด
􀂷 บวมแขนขา

การที่จะทราบแน่ชัดต้องทำการทดสอบภูมิแพ้ หลังจากที่ทราบชื่อยาที่แพ้แล้วก็จดชื่อยาที่แพ้ไว้กับตัว หรืออาจจะทำป้ายติดไว้กับตัว

การรักษาที่ดีที่สุดคือการหลีกเลี่ยงจากยาชนิดนั้นโดยเด็ดขาด ก่อนที่แพทย์จะจ่ายยาต้องบอกแพทย์ทุกครั้งว่าแพ้ยาอะไร



ก่อนรับยาจากเภสัชกรต้องถามชื่อยาและบอกว่าแพ้ยาอะไรแก่เภสัชกรเนื่องจากยาชนิดเดียวกันอาจจะมีหลายชื่อ ผลเสียที่เกิดจากยาอาจจะเกิดจากสาเหตุอื่น เช่น

􀂷 Overdose or toxicity ได้รับยาเกินขนาด เช่นการได้ยาปฏิชีวนะรักษาโรคติดเชื้อหากได้ติดต่อกันนานๆอาจจะมีผื่นเกิดขึ้น

􀂷 Secondary effects ผลข้างเคียงจากฤทธิ์ของยา เช่นเร่รับประทาน aspirin เพื่อแก้ปวดแต่เกิดเลือดออกง่าย เลือดออกง่ายเป็นผลจากยา aspirin

􀂷 Side effects คือผลข้างเคียงของยา เช่นกินยาลดน้ำมูกจะมีอาการปากแห้งใจสั่น นอนไม่หลับ กินยาแก้หอบหืดจะมีอาการมือสั่นใจสั่น กินยาแก้ปวดจะมีอาการปวดท้อง

ท่านสามารถอ่านผลข้างเคียงได้จากสลากยาที่กำกับ อาการข้างเคียงไม่จำเป็นต้องเกิดกับทุกคนที่กินยา อาจจะเกิดกับบางคนเท่านั้น



􀂷 Drug interactions
ท่านหากรับประทานยามากกว่าหนึ่งชนิดท่านต้องทราบว่ายาสองชนิดมีปฏิกิริยาส่งเสริมหรือหักล้างกันหรือไม่ทั้งในแง่ของการรักษาและผลข้างเคียงของยา
เช่นรับประทานยาชนิดหนึ่งและเมื่อได้ยาอีกชนิดหนึ่งซึ่งอาจจะส่งผลให้ยานั้นออกฤทธิ์หรือผลข้างเคียงมากขึ้นและอาจจะเป็นอันตรายแก่ผู้ใช้ยา

􀂷 Idiosyncratic reactions เป็นปฏิกิริยาที่เกิดโดยคาดการณ์ไม่ได้ว่าจะเกิดอาการภูมิแพ้หรือไม่
โปรดจำไว้ว่าหากท่านจะรับประทานยาหรือสมุนไพรท่านต้องคำนึงถึงการแพ้ยาทั้งที่เกิดจากปฏิกิริยาภูมิแพ้และจากอย่างอื่น นอกจากนั้นท่านที่รับประทานยามากกว่า
สองชนิดต้องระวังว่าอาจจะเกิดผลเสียแก่ตัวท่าน
 
การเกิดปฏิกิริยาภูมิแพ้

เมื่อร่างกายได้รับสารชนิดหนึ่งเข้าสู่ร่างกาย ทางผิวหนัง ทางรับประทาน ทางลมหายใจ หรือจากการฉีดยา หากร่งกายรับสารนั้นได้ก็ไม่เกิดผลเสียต่อร่างกาย
แต่หากสารนั้นเป็นสารที่ทำให้เกิดภูมิแพ้ก็จะทำให้เกิดผลเสียต่อร่างกาย อาจจะรุ่นแรงมากจนทำให้เกิดเสียชีวิตกระทันหัน เรียกว่ายังไม่ได้ถอนเข็มก็เกิดอาการแล้ว
หรือบางกรณีอาจจะเกิดปฏิกิริยาภูมิมาในภายหลังโดยที่ตัวคนไข้ไม่รู้ด้วยซ้ำว่าไปรับสารใดมาบ้าง
ความรุนแรงและความรวดเร็วของการเกิดภูมแพ้ขึ้นชนิดของภูมิแพ้ซึ่งแบ่งออกเป็น
IgE - Mediated Reaction
เมื่อร่างกายได้รับสารภูมิแพ้ร่างกายจะสร้างภูมิคุ้มกันชนิด IgE ขึ้นเพื่อกำจัดสิ่งแปลกปลอม
􀂷 IgE จะจับกับโปรตีนของสารภูมแพ้และเกาะกับผิวของเซลล์เม็ดเลือดขาวที่เรียกว่า Mast cell
􀂷 หลังจากนั้นจะเกิดปฏิกิริยาภูมิแพ้ตามมาทำให้เกิดการหลังของสารเคมอีกหลายชนิด histamine heparin Protease Eosinophil chemotactic factor Neutrophil chemotactic
factor ,Leucotriene ,prostaglandin
สารต่างๆเหล่านี้จะทำให้เกิดปฏิกิริยาภูมแพ้เฉียบพลันที่เรียกว่า Anaphylaxis ซึ่งมีอาการดังต่อไปนี้

􀂷 ลมพิษ
􀂷 ความดันโลหิตต่ำ
􀂷 คัน
􀂷 Angioedema
ตัวอย่างสารที่ทำให้เกิดภูมิแพ้ชนิดนี้
􀂷 ยาโดยเฉพาะกลุ่ม pennicillin
􀂷 การให้เลือด
􀂷 วัคซีน
􀂷 ฮอร์โมน

Cytotoxic/Cytolytic Reaction

ร่างกายจะสร้างภูมิชนิด IgG,iGm ,Complement มาจับกับโปรตีนของสารก่อภูมิแพ้ทำให้มีการทำลายของเซลล์โดยเฉพาะเซลล์ของเม็ดเลือดทำให้เกิด การแตกของเม็ดเลือดแดง(
Immune hemolytic anemia) เกล็ดเลือดต่ำ(Thromobocytopenia) เม็ดเลือดขาวต่ำ (Granulocytopenia) ตัวอย่างยาที่ทำให้เกิดปฏิกิริยาชนิดนี้
􀂷 pennicillin
􀂷 quinidine
􀂷 sulfonamide
􀂷 methyldopa
 

Immune Complex Reaction
ภูมิของร่างกายจะรวมกับโปรตีนของสารภูมิแพ้เกิดสารที่เรียกว่า immune complex ซึ่งจะไหลเวียนไปในกระแสเลือด เมื่อimmune complex
นี้ไปเกาะที่เส้นเลือดก็จะก่อให้เกิดปฏิกิริยาดังนี้
􀂷 เกร็ดเลือดจะมาเกาะรวมกลุ่ม plattlet aggregation
􀂷 มีการกระตุ้นเซลล์ Mast cell activation
􀂷 มีการกระตุ้น ทำให้เกิด การรั่วของผนังหลอดเลือด{permiability} การหลั่งสารที่ทำให้เกิดการอักเสบ(ทำให้เกิดปวด บวม แดง ร้อน)
อาการของภูมิแพ้ชนิดนี้ได้แก่
􀂷 ไข้
􀂷 ผื่นที่ผิวหนัง
􀂷 ต่อมน้ำเหลืองโต
􀂷 ปวดข้อ
􀂷 ไตอักเสบ
􀂷 ตับอักเสบ
ยาที่ทำให้เกิดปฏิกิริยาภูมิแพ้ชนิดนี้ได้แก่
􀂷 hydralazine ยาลดความดันโลหิต
􀂷 procanamide
􀂷 isoniazid ยารักษาวัณโรค
􀂷 phenyltoin ยากันชัก
T-cell Mediated Reaction
ปฏิกิริยาภูมิแพ้เกิดจากเซลลT-cell์ lymphocyte ถูกกระตุ้นเมื่อได้รับสารภูมิแพ้ อาการที่สำคัญของการเกิดภูมิแพ้ชนิดนี้คือพวกผื่นแพ้ที่เกิดจากการสัมผัส

แพ้ยาทำให้เกิดไข้

ท่านที่เป็นโรคติดเชื้อและซื้อยารับประทาน หลังจากรับประทานไประยะหนึ่งไข้ไม่ลง ซึ่งอาจจะเกิดจากแพ้ยาก็ได้ ยาที่เกิดอาจจะเป็นไข้ต่ำๆตลอด
หรือไข้สูงเป็นช่วงๆยาที่มักจะทำให้เกิดไข้คือยากลุ่มปฏิชีวนะ เมื่อหยุดยา 24-48 ชั่วโมงไข้ก็จะลงเอง
ยาที่ทำให้เกิดผลภูมิแพ้ที่ตับ
ปฏิกิริยาภูมิแพ้อาจจะทำให้เกิดการอักเสบของตับ โดยตับจะโตและเจ็บเมื่อเจาะเลือดตรวจจะพบว่ามีค่า SGOT,SGPT สูงและอาจจะมีดีซ่าน

ยาที่ทำให้เกิดตับอักเสบที่พบบ่อยได้แก่
􀂷 phenotiazine
􀂷 sulfonamide
􀂷 halathane
􀂷 phenyltoin
􀂷 Isoniazid

ยาที่ทำให้เกิดโรคปอด
ผู้ป่วยที่ใช้ยาเป็นประจำเช่นยา nitrofurantoin sulfasalaxine นานๆอาจะทำให้เกิดโรคที่ปอด ทำให้เกิด ไข้ ไอ และมีผื่น เมื่อเจาะเลือดพบว่า eosinphil ในเลือดสูง
การรักษาให้หยุดยานั้นเสีย

การแพ้ยา penicillin
ยากลุ่ม penicillin เป็นยาที่แพ้ได้บ่อยที่สุด การเกิดภูมิแพ้ได้หลายแบบ IgE,Immune Complex,Cytotoxic เรื่องน่ารู้เกี่ยวกับแพ้ยาpenicillin อาการแพ้มีได้หลายแบบ
􀂷 ลมพิษ
􀂷 คัน
􀂷 ผื่นได้หลายๆแบบ
􀂷 แพ้แบบรุนแรงได้แก่ หนังตา ปากบวมที่เรียกว่า angioedema กล่องเสียงบวม(laryngeal edema) หลอดลมเกร็ง()ความดันโลหิตต่ำ
􀂷 บางรายผื่นเป็นมากทำให้เกิดลอกทั้งตัวที่เรียกว่า steven johnson syndrome
􀂷 ในทางห้องทดลองพบว่าผู้ที่แพ้penicillin สามารถแพ้ยากลุ่ม cephalosporin ดังนั้นหากสามารกเลือกยากลุ่มอื่นได้น่าจะเป็นการปลอดภัย
􀂷 การแพ้ยา cephalosporin ก็ไม่จำเป็นต้องแพ้ penicillin

penicillin เมื่อให้ในโรคต่อไปนี้จะทำให้เกิดผื่นได้ง่าย

􀂷 ติดเชื้อไวรัสโดยเฉพาะ infectiuos mononucleosis
􀂷 ,มะเร็งเม็ดเลือดขาว
􀂷 กรดยูริกในเลือดสูง
􀂷 ให้ยาpenicillin ร่วมกับ allopurinol

แพ้ยา sulfonamide (เรียกติดปากว่าซัลฟา)

ยา sulfonamide เป็นยาผสมในยาหลายชนิดได้แก่ ยาปฏิชีวนะ(bactrim) ยาแก้ปวด() ยาขับปัสสาวะ ยาลดน้ำตาลในเลือด

การแพ้ aspirin
อาการของผู้ที่แพ้ aspirin มีได้หลายรูปแบบ
􀂷 ผื่นลมพิษ
􀂷 angioedema หน้าหนังจาปากบวม
􀂷 น้ำมูกไหล
􀂷 หลอดลมเกร็งทำให้แน่นหน้าอก หายใจไม่ออก หากเป็นมากอาจจะมีตัวเขียว ริมฝีปากเขียว
􀂷 ความดันโลหิตต่ำ
􀂷 ผู้ที่เป็นโรคหอบหืด หรือไซนัสอักเสบจะแพ้ได้ถึงร้อยละ30-40

การรักษาอาการแพ้ยา
สำหรับผู้ทีอาการแพ้เฉียบพลัน


􀂷 ให้หยุดยานั้นทันที
􀂷 หากมีอาการแพ้รุนแรงแบบ anaphylaxis ซึ่งอาจจะเป็นอันตรายถึงกับชีวิต ต้องได้รับยา epinephrine
􀂷 สำหรับผู้ที่มีผื่นลมพิษหรือ angioedema ให้ยาแก้แพ้รับประทาน
􀂷 ให้ยา steroid ชนิดรับประทาน

สำหรับผู้ที่แพ้ไม่เฉียบพลัน

􀂷 ให้หยุดยาที่สงสัย หลังหยุดยาผื่นอาจจะยังเกิดขึ้นต่อไปได้อีก
􀂷 หากผื่นเป็นน้อยให้ยาแก้แพ้ชนิดเดียวก็น่าจะพอ
􀂷 สำหรับผู้ที่มีผื่นมากและมีท่าจะเป็นมากขึ้นก็สามารถให้กิน steroid ชนิดกินระยะสั่นๆ
􀂷 สำหรับผู้ที่มีการอักเสบของไต serum sickness ปวดข้อ อาจจะต้องให้ยา steroid และยาแก้แพ้รับประทาน